13.07.2015 Views

zoonoses and communicable diseases common to ... - PAHO/WHO

zoonoses and communicable diseases common to ... - PAHO/WHO

zoonoses and communicable diseases common to ... - PAHO/WHO

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

ZOONOSES AND COMMUNICABLE DISEASESCOMMON TO MAN AND ANIMALSThird EditionVolume IBacterioses <strong>and</strong> MycosesScientific <strong>and</strong> Technical Publication No. 580PAN AMERICAN HEALTH ORGANIZATIONPan American Sanitary Bureau, Regional Office of theWORLD HEALTH ORGANIZATION525 Twenty-third Street, N.W.Washing<strong>to</strong>n, D.C. 20037 U.S.A.2001


Also published in Spanish (2001) with the title:Zoonosis y enfermedades transmisibles comunes al hombre a los animalesISBN 92 75 31580 9<strong>PAHO</strong> Cataloguing-in-PublicationPan American Health OrganizationZoonoses <strong>and</strong> <strong>communicable</strong> <strong>diseases</strong> <strong>common</strong> <strong>to</strong> man <strong>and</strong> animals3rd ed. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>, © 2001.3 vol.—(Scientific <strong>and</strong> Technical Publication No. 580)ISBN 92 75 11580 XI. Title II. Series1. ZOONOSES2. BACTERIAL INFECTIONS AND MYCOSES3. COMMUNICABLE DISEASE CONTROL4. FOOD CONTAMINATION5. PUBLIC HEALTH VETERINARY6. DISEASE RESERVOIRSNLM WC950.P187 2001 EnThe Pan American Health Organization welcomes requests for permission <strong>to</strong>reproduce or translate its publications, in part or in full. Applications <strong>and</strong> inquiriesshould be addressed <strong>to</strong> the Publications Program, Pan American HealthOrganization, Washing<strong>to</strong>n, D.C., U.S.A., which will be glad <strong>to</strong> provide the latestinformation on any changes made <strong>to</strong> the text, plans for new editions, <strong>and</strong> reprints<strong>and</strong> translations already available.©Pan American Health Organization, 2001Publications of the Pan American Health Organization enjoy copyright protectionin accordance with the provisions of Pro<strong>to</strong>col 2 of the Universal CopyrightConvention. All rights are reserved.The designations employed <strong>and</strong> the presentation of the material in this publicationdo not imply the expression of any opinion whatsoever on the part of the Secretaria<strong>to</strong>f the Pan American Health Organization concerning the status of any country, terri<strong>to</strong>ry,city or area or of its authorities, or concerning the delimitation of its frontiersor boundaries.The mention of specific companies or of certain manufacturers’ products does notimply that they are endorsed or recommended by the Pan American HealthOrganization in preference <strong>to</strong> others of a similar nature that are not mentioned.Errors <strong>and</strong> omissions excepted, the names of proprietary products are distinguishedby initial capital letters.


CONTENTSPrologue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . viiPreface <strong>to</strong> the First Edition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ixPreface <strong>to</strong> the Second Edition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xiIntroduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xvPART I: BACTERIOSESActinomycosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3Aeromoniasis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6Animal Erysipelas <strong>and</strong> Human Erysipeloid . . . . . . . . . . . . . . . . . . . . . . . . . . 14Anthrax . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21Botulism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28Brucellosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40Campylobacteriosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67Cat-scratch Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78Clostridial Food Poisoning. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82Clostridial Wound Infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87Colibacillosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90Corynebacteriosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99Derma<strong>to</strong>philosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103Diseases Caused by Nontuberculous Mycobacteria . . . . . . . . . . . . . . . . . . . . 107Diseases in Man <strong>and</strong> Animals Caused by Non-O1 Vibrio cholerae. . . . . . . . . 117Enterocolitic Yersiniosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122Enterocolitis Due <strong>to</strong> Clostridium difficile . . . . . . . . . . . . . . . . . . . . . . . . . . . . 132Food Poisoning Caused by Vibrio parahaemolyticus . . . . . . . . . . . . . . . . . . . 138Gl<strong>and</strong>ers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142Infection Caused by Capnocy<strong>to</strong>phaga canimorsus <strong>and</strong> C. cynodegmi. . . . . . . 146Leprosy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149Lep<strong>to</strong>spirosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 157Listeriosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168Lyme Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179Melioidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184Necrobacillosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190Nocardiosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195Pasteurellosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199Plague . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207Pseudotuberculous Yersiniosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218Rat-bite Fever . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226Rhodococcosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 229Salmonellosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 233Shigellosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247Staphylococcal Food Poisoning . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251Strep<strong>to</strong>coccosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257Tetanus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265Tick-borne Relapsing Fever . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 271iii


ivCONTENTSTularemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 275Zoonotic Tuberculosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 283PART II: MYCOSESAdiaspiromycosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303Aspergillosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305Blas<strong>to</strong>mycosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 311C<strong>and</strong>idiasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315Coccidioidomycosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 320Cryp<strong>to</strong>coccosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 326Derma<strong>to</strong>phy<strong>to</strong>sis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 332His<strong>to</strong>plasmosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 339Myce<strong>to</strong>ma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 345Pro<strong>to</strong>thecosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 348Rhinosporidiosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 350Sporotrichosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 352Zygomycosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 356Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361LIST OF TABLES AND ILLUSTRATIONSBacteriosesTables1. Foods giving rise <strong>to</strong> botulism, <strong>and</strong> number of outbreaks,United States of America, 1899–1977 . . . . . . . . . . . . . . . . . . . . . . . . . . . 312. Number of cases <strong>and</strong> deaths from human plague in theAmericas, 1971–1980 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2103. Outbreaks of foodborne salmonellosis in selected countries,1981–1985 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2354. Distribution of tetanus morbidity according <strong>to</strong> political division<strong>and</strong> climate, Argentina, 1967–1977 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 267Figures1. Animal erysipelas <strong>and</strong> human erysipeloid (Erysipelothrixrhusiopathiae). Mode of transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . 172. Anthrax. Transmission cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253. Botulism (transmitted by foods). Reported cases <strong>and</strong> deathsper year, United States of America, 1960–1980 . . . . . . . . . . . . . . . . . . . . 294. Reported cases of botulism per year, Argentina, 1967–1981 . . . . . . . . . . 325. Bovine brucellosis (Brucella abortus). Mode of transmission . . . . . . . . . 526. Swine brucellosis (Brucella suis). Mode of transmission . . . . . . . . . . . . . 53


CONTENTSv7. Caprine <strong>and</strong> ovine brucellosis (Brucella melitensis).Mode of transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 548. Campylobacteriosis (Campylobacter jejuni). Mode of transmission. . . . . 709. Campylobacteriosis (Campylobacter fetus). Probable modeof transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7510. Enterocolitic yersiniosis (Yersinia enterocolitica). Supposedmode of transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12711. Gl<strong>and</strong>ers. Mode of transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14412. Lep<strong>to</strong>spirosis. Synanthropic transmission cycle . . . . . . . . . . . . . . . . . . . . 16213. Melioidosis (Pseudomonas pseudomallei). Mode of transmission . . . . . . 18714. Number of cases <strong>and</strong> deaths from human plague worldwide,1971–1980 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20915. Plague. Domestic <strong>and</strong> peridomestic transmission cycle . . . . . . . . . . . . . . 21316. Pseudotuberculous yersiniosis (Yersinia pseudotuberculosis).Probable mode of transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22217. Salmonellosis. Mode of transmission (except Salmonella typhi<strong>and</strong> the paratyphoid serotypes) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24118. Tick-borne relapsing fever (Ornithodoros spp.).Mode of transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27319. Tularemia. Mode of transmission in the Americas . . . . . . . . . . . . . . . . . . 27920. Tuberculosis (Mycobacterium bovis). Mode of transmission . . . . . . . . . . 293


PROLOGUEZoonoses <strong>and</strong> <strong>communicable</strong> <strong>diseases</strong> <strong>common</strong> <strong>to</strong> man <strong>and</strong> animals continue <strong>to</strong>have high incidence rates <strong>and</strong> <strong>to</strong> cause significant morbidity <strong>and</strong> mortality.Infections <strong>and</strong> parasi<strong>to</strong>ses of cattle can reduce meat or milk production <strong>and</strong> can lead<strong>to</strong> the death or destruction of the animals, all of which diminishes the supply ofavailable food for man. These <strong>diseases</strong> are also an obstacle for international trade,as well as a serious financial drain for cattle farmers <strong>and</strong>, more broadly, for a community’sor a country’s economy, which can have wide repercussions for a society’shealth.With the aim of helping <strong>to</strong> solve these problems, the Pan American HealthOrganization (<strong>PAHO</strong>)—an international public health organization that has devoteditself <strong>to</strong> improving the health <strong>and</strong> living conditions of the people of the Americas fornearly one hundred years—established the Veterinary Public Health Program. TheProgram’s overall objective is <strong>to</strong> collaborate with <strong>PAHO</strong>’s Member Countries in thedevelopment, implementation, <strong>and</strong> evaluation of policies <strong>and</strong> programs that lead <strong>to</strong>food safety <strong>and</strong> protection <strong>and</strong> <strong>to</strong> the prevention, control, or eradication of <strong>zoonoses</strong>,among them foot-<strong>and</strong>-mouth disease.To this end, <strong>PAHO</strong>’s Veterinary Public Health Program has two specializedregional centers: the Pan American Foot-<strong>and</strong>-Mouth Disease Center (PANAFTOSA),created in 1951 in Rio de Janeiro, Brazil, <strong>and</strong> the Pan American Institute for FoodProtection <strong>and</strong> Zoonoses (INPPAZ), established on November 15, 1991 in BuenosAires, Argentina. INPPAZ’s precursor was the Pan American Zoonoses Center(CEPANZO), which was created through an agreement with the Government ofArgentina <strong>to</strong> help the countries of the Americas combat <strong>zoonoses</strong>, <strong>and</strong> which operatedfrom 1956 until 1990.Since its creation in 1902, <strong>PAHO</strong> has participated in various technical cooperationactivities with the countries, among them those related <strong>to</strong> the surveillance, prevention,<strong>and</strong> control of <strong>zoonoses</strong> <strong>and</strong> <strong>communicable</strong> <strong>diseases</strong> <strong>common</strong> <strong>to</strong> man <strong>and</strong>animals, which cause high morbidity, disability, <strong>and</strong> mortality in vulnerable humanpopulations. <strong>PAHO</strong> has also collaborated in the strengthening of preventive medicine<strong>and</strong> public health through the promotion of veterinary health education in learning,research, <strong>and</strong> health care centers. An example of this work is the preparation ofseveral publications, among which the two previous Spanish <strong>and</strong> English editions ofZoonoses <strong>and</strong> Communicable Diseases Common <strong>to</strong> Man <strong>and</strong> Animals st<strong>and</strong> out.Scientific knowledge has progressed since the last edition. Also, the countries ofthe Americas have modified their lives<strong>to</strong>ck production strategies in recent years,which has affected the transmission of zoonotic infections <strong>and</strong> their distribution. Thepublication of this third edition is an attempt <strong>to</strong> address these changes. The third editionis presented in three volumes: the first contains bacterioses <strong>and</strong> mycoses; thesecond, chlamydioses, rickettsioses, <strong>and</strong> viroses; <strong>and</strong> the third, parasi<strong>to</strong>ses.We believe that this new edition will continue <strong>to</strong> be useful for professors <strong>and</strong> studentsof public health, medicine, <strong>and</strong> veterinary medicine; workers in public health<strong>and</strong> animal health institutions; <strong>and</strong> veterinarians, researchers, <strong>and</strong> others interestedin the subject. We also hope that this publication is a useful <strong>to</strong>ol in the elaborationof national zoonosis control or eradication policies <strong>and</strong> programs, as well as in riskvii


viiiPROLOGUEevaluation <strong>and</strong> in the design of epidemiological surveillance systems for theprevention <strong>and</strong> timely control of emerging <strong>and</strong> reemerging <strong>zoonoses</strong>. In summary,we are confident that this book will contribute <strong>to</strong> the application of the knowledge<strong>and</strong> resources of the veterinary sciences for the protection <strong>and</strong> improvement ofpublic health.GEORGE A.O. ALLEYNEDIRECTOR


xPREFACE TO THE FIRST EDITIONgiven the appropriate ecologic fac<strong>to</strong>rs for existence of the etiologic agents. Today,public health <strong>and</strong> animal health administra<strong>to</strong>rs, physicians, <strong>and</strong> veterinarians mustbe familiar with the geographic distribution <strong>and</strong> pathologic manifestations of thevarious infectious agents so that they can recognize <strong>and</strong> prevent the introduction ofexotic <strong>diseases</strong>.We, the authors, would like <strong>to</strong> give special recognition <strong>to</strong> Dr. Joe R. Held,Assistant Surgeon-General of the United States Public Health Service <strong>and</strong> Direc<strong>to</strong>rof the Division of Research Services of the U.S. National Institutes of Health, whogave impetus <strong>to</strong> the English translation <strong>and</strong> reviewed the bacterioses sections.We would also like <strong>to</strong> express our utmost appreciation <strong>to</strong> the experts whoreviewed various portions of this book <strong>and</strong> offered their suggestions for improvingthe text. These include: Dr. Jeffrey F. Williams, Professor in the Department ofMicrobiology <strong>and</strong> Public Health, Michigan State University, who reviewed thechapters dealing with parasitic <strong>zoonoses</strong>; Dr. James Bond, <strong>PAHO</strong>/<strong>WHO</strong> RegionalAdviser in Viral Diseases, who read the viroses; Dr. An<strong>to</strong>nio Pío, formerly<strong>PAHO</strong>/<strong>WHO</strong> Regional Adviser in Tuberculosis <strong>and</strong> presently with <strong>WHO</strong> in Geneva,<strong>and</strong> Dr. James H. Rust, <strong>PAHO</strong>/<strong>WHO</strong> Regional Adviser in Enteric Diseases, both ofwhom reviewed the bacterioses; <strong>and</strong> Dr. F. J. López Antuñano, <strong>PAHO</strong>/<strong>WHO</strong>Regional Adviser in Parasitic Diseases, who read the metazooses.We would like <strong>to</strong> thank Dr. James Cocozza, <strong>PAHO</strong>/<strong>WHO</strong> Veterinary Adviser, forhis review of the translation <strong>and</strong> Dr. Judith Navarro, Edi<strong>to</strong>r in the Office ofPublications of <strong>PAHO</strong>, for her valuable collaboration in the edi<strong>to</strong>rial revision <strong>and</strong>composition of the book.PEDRO N. ACHABORIS SZYFRES


PREFACE TO THE SECOND EDITIONThe fine reception accorded the Spanish, English, <strong>and</strong> French versions of thisbook has motivated us <strong>to</strong> revise it in order that it still may serve the purpose forwhich it was written: <strong>to</strong> provide an up-<strong>to</strong>-date source of information <strong>to</strong> the medicalprofession <strong>and</strong> allied fields. This book has undoubtedly filled a void, judging by itswide use in schools of public health, medicine, <strong>and</strong> veterinary medicine, as well asby bureaus of public <strong>and</strong> animal health.The present edition has been considerably enlarged. In the seven years since thefirst edition was published, our knowledge of <strong>zoonoses</strong> has increased broadly <strong>and</strong>rapidly, <strong>and</strong> new zoonotic <strong>diseases</strong> have emerged. Consequently, most of the discussionshave been largely rewritten, <strong>and</strong> 28 new <strong>diseases</strong> have been added <strong>to</strong> theoriginal 148. Some of these new <strong>diseases</strong> are emerging <strong>zoonoses</strong>; others are pathologicentities that have been known for a long time, but for which the epidemiologicconnection between man <strong>and</strong> animal has been unclear until recently.The use this book has had outside the Western Hemisphere has caused us <strong>to</strong> ab<strong>and</strong>onthe previous emphasis on the Americas in favor of a wider scope <strong>and</strong> geomedicalview. Moreover, wars <strong>and</strong> other conflicts have given rise <strong>to</strong> the migration ofpopulations from one country or continent <strong>to</strong> another. A patient with a diseasehere<strong>to</strong>fore known only in Asia may now turn up in Amsterdam, London, or NewYork. The physician must be aware of these <strong>diseases</strong> in order <strong>to</strong> diagnose <strong>and</strong> treatthem. “Exotic” animal <strong>diseases</strong> have been introduced from Africa <strong>to</strong> Europe, theCaribbean, <strong>and</strong> South America, causing great damage. The veterinary physicianmust learn <strong>to</strong> recognize them <strong>to</strong> be able <strong>to</strong> prevent <strong>and</strong> eradicate them before theybecome entrenched. It must be remembered that parasites, viruses, bacteria, <strong>and</strong>other agents of zoonotic infection can take up residence in any terri<strong>to</strong>ry where theyfind suitable ecologic conditions. Ignorance, economic or personal interests, <strong>and</strong>human cus<strong>to</strong>ms <strong>and</strong> needs also favor the spread of these <strong>diseases</strong>.Research in recent years has demonstrated that some <strong>diseases</strong> previously considered<strong>to</strong> be exclusively human have their counterparts in wild animals, which in certaincircumstances serve as sources of human infection. On the other h<strong>and</strong>, theseanimals may also play a positive role by providing models for research, such as inthe case of natural leprosy in nine-b<strong>and</strong>ed armadillos or in nonhuman primates inAfrica. Of no less interest is the discovery of Rickettsia prowazekii in eastern flyingsquirrels <strong>and</strong> in their ec<strong>to</strong>parasites in the United States, <strong>and</strong> the transmission of theinfection <strong>to</strong> man in a country where epidemic typhus has not been seen since 1922.A possible wild cycle of dengue fever is also discussed in the book. Is Creutzfeldt-Jakob disease a zoonosis? No one can say with certainty, but some researchersbelieve it may have originated as such. In any case, interest is aroused by the surprisingsimilarity of this disease <strong>and</strong> of kuru <strong>to</strong> animal subacute spongiformencephalopathies, especially scrapie, the first known <strong>and</strong> best studied of this group.Discussion of human <strong>and</strong> animal slow viruses <strong>and</strong> encephalopathies is included inthe spirit of openness <strong>to</strong> possibilities <strong>and</strong> the desire <strong>to</strong> bring the experience of onefield of medicine <strong>to</strong> another. In view of worldwide concern over acquired immunodeficiencysyndrome (AIDS), a brief section on retroviruses has also been added, inwhich the relationship between the human disease <strong>and</strong> feline <strong>and</strong> simian AIDS isxi


xiiPREFACE TO THE SECOND EDITIONnoted. Another <strong>to</strong>pic deeply interesting <strong>to</strong> researchers is the mystery of the radicalantigenic changes of type A influenza virus, a cause of explosive p<strong>and</strong>emics thataffect millions of persons around the world. Evidence is mounting that thesechanges result from recombination with a virus of animal origin (see Influenza).That this should occur is not surprising, given the constant interaction between man<strong>and</strong> animals. As a rule, <strong>zoonoses</strong> are transmitted from animal <strong>to</strong> man, but the reversemay also occur, as is pointed out in the chapters on hepatitis, herpes simplex, <strong>and</strong>measles. The victims in these cases are nonhuman primates, which may in turnretransmit the infection <strong>to</strong> man under certain circumstances.Among emerging <strong>zoonoses</strong> we cite Lyme disease, which was defined as a clinicalentity in 1977; the etiologic agent was found <strong>to</strong> be a spirochete (isolated in 1982),for which the name Borrelia burgdorferi was recently proposed. Emerging viral<strong>zoonoses</strong> of note in Latin America are Rocio encephalitis <strong>and</strong> Oropouche fever; thelatter has caused multiple epidemics with thous<strong>and</strong>s of victims in northeast Brazil.Outst<strong>and</strong>ing among new viral disease problems in Africa are the emergence of Eboladisease <strong>and</strong> the spread of Rift Valley fever virus, which has caused tens of thous<strong>and</strong>sof human cases along with great havoc in the cattle industry of Egypt <strong>and</strong> has evokedalarm around the world. Similarly, the pro<strong>to</strong>zoan Cryp<strong>to</strong>sporidium is emerging asone of the numerous agents of diarrheal <strong>diseases</strong> among man <strong>and</strong> animals, <strong>and</strong> probablyhas a worldwide distribution.As the English edition was being prepared, reports came <strong>to</strong> light of two animal<strong>diseases</strong> not previously confirmed in humans. Three cases of human pseudorabiesvirus infection were recognized between 1983 <strong>and</strong> 1986 in two men <strong>and</strong> one womanwho had all had close contact with cats <strong>and</strong> other domestic animals. In 1986, serologictesting confirmed infection by Ehrlichia canis in a 51-year-old man who hadbeen suspected of having Rocky Mountain spotted fever. This is the first knownoccurrence of E. canis infection in a human. These two <strong>diseases</strong> bear watching aspossible emerging <strong>zoonoses</strong>.The space given <strong>to</strong> each zoonosis is in proportion <strong>to</strong> its importance. Some <strong>diseases</strong>that deserve their own monographs were given more detailed treatment, but noattempt was made <strong>to</strong> cover the <strong>to</strong>pic exhaustively.We, the authors, would like <strong>to</strong> give special recognition <strong>to</strong> Dr. Donald C. Blenden,Professor in the Department of Medicine <strong>and</strong> Infectious Diseases, School ofMedicine, <strong>and</strong> Head of the Department of Veterinary Microbiology, College ofVeterinary Medicine, University of Missouri; <strong>and</strong> <strong>to</strong> Dr. Manuel J. Torres, Professorof Epidemiology <strong>and</strong> Public Health, Department of Veterinary Microbiology,College of Veterinary Medicine, University of Missouri, for their thorough review of<strong>and</strong> valuable contributions <strong>to</strong> the English translation of this book.We would also like <strong>to</strong> recognize the support received from the Pan AmericanHealth Organization (<strong>PAHO</strong>/<strong>WHO</strong>), the Pan American Health <strong>and</strong> EducationFoundation (PAHEF), <strong>and</strong> the Pan American Zoonoses Center in Buenos Aires,Argentina, which enabled us <strong>to</strong> update this book.We are most grateful <strong>to</strong> Dr. F. L. Bryan for his generous permission <strong>to</strong> adapt hismonograph “Diseases Transmitted by Foods” as an Appendix <strong>to</strong> this book.


PREFACE TO THE SECOND EDITIONxiiiMr. Carlos Larranaga, Chief of the Audiovisual Unit at the Pan AmericanZoonosis Center, deserves our special thanks for the book’s artwork, as do Ms. IrisElliot <strong>and</strong> Mr. William A. Stapp for providing the translation in<strong>to</strong> English. We wouldlike <strong>to</strong> express our most sincere gratitude <strong>and</strong> recognition <strong>to</strong> Ms. Donna J. Reynolds,edi<strong>to</strong>r in the <strong>PAHO</strong> Edi<strong>to</strong>rial Service, for her valuable collaboration in the scientificedi<strong>to</strong>rial revision of the book.PEDRO N. ACHABORIS SZYFRES


INTRODUCTIONThis new edition of Zoonoses <strong>and</strong> Communicable Diseases Common <strong>to</strong> Man <strong>and</strong>Animals is published in three volumes: I. Bacterioses <strong>and</strong> mycoses; II.Chlamydioses <strong>and</strong> rickettsioses, <strong>and</strong> viroses; <strong>and</strong> III. Parasi<strong>to</strong>ses. Each of the fiveparts corresponds <strong>to</strong> the location of the etiologic agents in the biological classification;for practical purposes, chlamydias <strong>and</strong> rickettsias are grouped <strong>to</strong>gether.In each part, the <strong>diseases</strong> are listed in alphabetical order <strong>to</strong> facilitate readersearches. There is also an alphabetical index, which includes synonyms of the <strong>diseases</strong><strong>and</strong> the etiologic agents’ names.In this edition, the numbers <strong>and</strong> names of the <strong>diseases</strong> according <strong>to</strong> theInternational Statistical Classification of Diseases <strong>and</strong> Related Health Problems,Tenth Revision (ICD-10), are listed below the disease title. However, some <strong>zoonoses</strong>are not included in ICD-10 <strong>and</strong> are difficult <strong>to</strong> classify within the current scheme.In addition, for each disease or infection, elements such as synonyms; etiology;geographical distribution; occurrence in man <strong>and</strong> animals; the disease in man <strong>and</strong>animals; source of infection <strong>and</strong> mode of transmission; role of animals in the epidemiology;diagnosis; <strong>and</strong> control are addressed. Patient treatment (for man or otherspecies) is beyond the scope of this work; however, recommended medicines areindicated for many <strong>diseases</strong>, especially where they are applicable <strong>to</strong> prophylaxis.Special attention is paid <strong>to</strong> the epidemiological <strong>and</strong> ecological aspects so that thereader can begin <strong>to</strong> underst<strong>and</strong> the determining fac<strong>to</strong>rs of the infection or disease.Some <strong>to</strong>pics include simple illustrations of the etiologic agent’s mode of transmission,showing the animals that maintain the cycle of infection in nature. Similarly,other graphics <strong>and</strong> tables are included <strong>to</strong> provide additional information on the geographicaldistribution or prevalence of certain <strong>zoonoses</strong>.The data on the occurrence of the infection in man <strong>and</strong> animals, along with dataon the geographical distribution, may help the reader judge the relative impact thateach disease has on public health <strong>and</strong> the lives<strong>to</strong>ck economy in the different regionsof the world, given that the importance of different <strong>zoonoses</strong> varies greatly. Forexample, foot-<strong>and</strong>-mouth disease is extremely important from an economic st<strong>and</strong>point,but of little importance in terms of public health, if animal protein losses arenot considered. In contrast, Argentine <strong>and</strong> Bolivian hemorrhagic fevers are importanthuman <strong>diseases</strong>, but their economic impact is minimal, if treatment costs <strong>and</strong>loss of man-hours are not taken in<strong>to</strong> account. Many other <strong>diseases</strong>, such as brucellosis,lep<strong>to</strong>spirosis, salmonellosis, <strong>and</strong> equine encephalitis, are important from botha public health <strong>and</strong> an economic st<strong>and</strong>point.Finally, each disease entry includes an alphabetical bibliography, which includesboth the works cited <strong>and</strong> other relevant works that the reader may consult for moreinformation about the disease.xv


ACTINOMYCOSISICD-10 A42.9Synonyms: Actinostrep<strong>to</strong>trichosis, m<strong>and</strong>ibular cancer, ray fungus disease.Etiology: Actinomyces israelii is the principal etiologic agent in man, <strong>and</strong> A.bovis the main one in animals. A. naeslundi, A. viscosus, A. odon<strong>to</strong>lytical, A. meyeri<strong>and</strong> Arachnia propionica (A. propionicus) are isolated less often, although A. viscosusplays an important role in canine actinomycosis. Some reports indicate isolationof A. israelii from animals (Georg, 1974) <strong>and</strong> A. bovis from man (Brunner et al.,1973). Actinomyces are higher bacteria with many characteristics of fungi. They aregram-positive, do not produce spores, are non–acid-fast, range from anaerobic <strong>to</strong>microaerophilic, <strong>and</strong> are part of the normal flora of the mouth <strong>and</strong> of women’s genitaltract (Burden, 1989).Geographic Distribution: Worldwide.Occurrence in Man: Infrequent; however, data are very limited. Fewer than 100cases of the disease are recorded each year by the Public Health Labora<strong>to</strong>ryService’s Communicable Disease Surveillance Centre in Great Britain (Burden,1989). According <strong>to</strong> older data, 368 cases were recorded in Wales <strong>and</strong> Engl<strong>and</strong> over12 years (1957–1968), with an incidence of 0.665 per million inhabitants, with ahigher incidence among industrial workers (Wilson, 1984). In Scotl<strong>and</strong>, the annualincidence was three per million <strong>and</strong> the rate of attack was 10 times higher in agriculturalworkers than among others.The his<strong>to</strong>rical ratio of two cases in men <strong>to</strong> one in women is probably no longervalid because of the number of cases of genital actinomycosis in women usingintrauterine contraceptive devices (IUDs).Occurrence in Animals: The frequency of the disease varies widely amongregions <strong>and</strong> is also influenced by different lives<strong>to</strong>ck management practices. The diseaseusually appears as sporadic cases. Small outbreaks have occurred in somemarshy areas of the United States <strong>and</strong> the former Soviet Union.The Disease in Man: A. israelii, the main causal agent in man, is a normal componen<strong>to</strong>f the flora of the mouth. As a result of wounds or surgery, it can enter thesoft tissues <strong>and</strong> bones, where it causes a suppurative granuloma<strong>to</strong>us process tha<strong>to</strong>pens <strong>to</strong> the surface through fistulas. Several clinical forms have been identifiedaccording <strong>to</strong> their location: cervicofacial, thoracic, abdominal, <strong>and</strong> generalized.Cervicofacial, which is the most <strong>common</strong> (from 50% <strong>to</strong> more than 70% of cases), isusually caused by a <strong>to</strong>oth extraction or a jaw injury; it begins with a hard swellingunder the mucous membrane of the mouth, beneath the periosteum of the m<strong>and</strong>ible,or in the skin of the neck. At a later stage, softened areas, depressions, <strong>and</strong> openings<strong>to</strong> the exterior with a purulent discharge are evident. These secretions usually containthe characteristic “sulphur granules,” which are actinomyces colonies. The thoracicform is generally caused by breathing the etiologic agent in<strong>to</strong> the bronchialtubes where it establishes a chronic bronchopneumonia that affects the lower portionsof the right lung (Burden, 1989), with symp<strong>to</strong>ms similar <strong>to</strong> pulmonary tuberculosis.As the disease progresses, invasion of the thoracic wall <strong>and</strong> its perforation3


4 BACTERIOSESby fistulous tracks may occur. The abdominal form usually occurs after surgery <strong>and</strong>appears as an encapsulated lesion that often becomes localized in the cecum <strong>and</strong> theappendix, where it produces hard tumors that adhere <strong>to</strong> the abdominal wall.The generalized form is infrequent <strong>and</strong> results from the erosive invasion of bloodvessels <strong>and</strong> lymphatic system, resulting in liver <strong>and</strong> brain disease.In recent years, reports of actinomycosis in the genital tract of women usingintrauterine contraceptive devices have multiplied, with the rate of infection increasingin proportion <strong>to</strong> the duration of IUD use. In one study (Valicenti et al., 1982),the infection was found in 1.6% of women in the general population of IUD users<strong>and</strong> in 5.3% of those attending the clinics. Another study of 478 IUD users found arate of infection of 12.6% based on Papanicolaou (Pap) smears (Koebler et al.,1983). Attempts <strong>to</strong> isolate the bacteria in Pap smears rarely yield positive results.However, A. israelii is also isolated from the genital tract of women who do not useIUDs, indicating that actinomyces are part of the normal flora (Burden, 1989). In thevast majority of cases, colonization by actinomyces produces only a superficial orasymp<strong>to</strong>matic infection.Treatment consists of prolonged high doses of penicillin (weeks or months).Erythromycin, clindamycin, <strong>and</strong> tetracycline may also be used. Surgical drainage ofabscesses is important. In women with an endometrium colonized by actinomyces,removing the IUD is sometimes enough for the endometrium <strong>to</strong> return <strong>to</strong> normal.The Disease in Animals: A. bovis is the principal agent of actinomycosis inbovines <strong>and</strong>, occasionally, in other animal species. In bovines, it centers chiefly inthe maxillae where it forms a granuloma<strong>to</strong>us mass with necrotic areas that developin<strong>to</strong> abscesses. These open via fistulous passages <strong>and</strong> discharge a viscous, odorless,yellow pus. The pus contains small, yellow, sulphur granules, which are rosetteshapedwhen viewed under a microscope. In some cases chewing becomes very difficult,<strong>and</strong> the animal s<strong>to</strong>ps eating <strong>and</strong> loses weight.The cost-benefit ratio must be measured when treating bovine <strong>and</strong> equine actinomycosis.Long-st<strong>and</strong>ing chronic lesions do not respond readily <strong>to</strong> treatment. If thelesions are small <strong>and</strong> circumscribed, they may be removed surgically. In other cases,curettage can be performed on the abscesses <strong>and</strong> fistulas, which are then packedwith gauze saturated with iodine tincture. Medical treatment is the same as forhuman actinomycosis, preferably using penicillin.In swine the etiologic agent localizes principally in the sow’s udder, where it givesrise <strong>to</strong> abscesses <strong>and</strong> fistulas. Its pathway of penetration is the lesion caused by theteeth of suckling pigs. This infection is attributed <strong>to</strong> Actinomyces suis, whose taxonomyis still uncertain.In dogs, the disease produces cervicofacial abscesses, empyemas accompanied bypleurisy <strong>and</strong> osteomyelitis, <strong>and</strong>, more rarely, abdominal abscesses <strong>and</strong> cutaneousgranulomas. The most <strong>common</strong> agent encountered prior <strong>to</strong> 1982 was A. viscosus(Hardie <strong>and</strong> Barsanti, 1982).Source of Infection <strong>and</strong> Mode of Transmission: The infection is endogenous.Actinomyces develop as saprophytes within <strong>and</strong> around carious teeth, in the mucinon dental enamel <strong>and</strong> in the <strong>to</strong>nsillar crypts. In studies carried out in several countries,actinomyces have been found in 40% of excised <strong>to</strong>nsils <strong>and</strong> have been isolatedin 30% <strong>to</strong> 48% of saliva samples or material from decayed teeth, as well asfrom the vaginal secretions of 10% of women using IUDs (Benenson, 1992).


ACTINOMYCOSIS 5Infections <strong>and</strong> pathological developments are the product of tissue trauma, lesions,or prolonged irritation. It has not been possible <strong>to</strong> isolate the agent of actinomycosisfrom the environment. It is believed that the causal agent penetrates the tissuesof the mouth through lesions caused by foods or foreign objects, or by way of dentaldefects. From the oral cavity, the bacteria can be swallowed or breathed in<strong>to</strong> thebronchial tubes.Role of Animals in the Epidemiology of the Disease: The species ofActinomyces that attack man are different from those that affect animals. Rarely isA. israelii found in animals or A. bovis found in man. The designation of speciesprior <strong>to</strong> 1960 is doubtful (Lerner, 1991) <strong>and</strong> thus, distinguishing one species fromanother presents great problems. The infection in animals is not transmitted <strong>to</strong> man,nor is it transmitted from person <strong>to</strong> person or animal <strong>to</strong> animal.Diagnosis: The clinical picture may be confused with other infections, such asactinobacillosis, nocardiosis, <strong>and</strong> staphylococcosis, as well as neoplasia <strong>and</strong> tuberculosis.The first step in confirming the diagnosis is <strong>to</strong> obtain pus, sputum, or tissuesamples for microscopic examination <strong>and</strong> culture, <strong>and</strong> <strong>to</strong> inspect them for granules.Filament masses are visible by direct observation. In smears of crushed granules orpus stained by the Gram <strong>and</strong> Kinyoun methods, gram-positive <strong>and</strong> non–acid-fastfilaments or pleomorphic forms, occasionally with bacillary-sized branching, maybe seen (Cottral, 1978). It is possible <strong>to</strong> identify the species of actinomyces causingthe disease only by culturing <strong>and</strong> typing the isolated microorganism. In testingwomen who use IUDs, direct immunofluorescence has yielded good results(Valicenti et al., 1982).Control: Prevention in man consists of proper oral hygiene <strong>and</strong> care after dentalextractions or other surgery in the oral cavity. No practical means have been establishedyet <strong>to</strong> prevent actinomycosis in animals.BibliographyAjello, L., L.K. Georg, W. Kaplan, L. Kaufman. Labora<strong>to</strong>ry Manual for MedicalMycology. Washing<strong>to</strong>n, D.C.: U.S. Government Printing Office; 1963. (Public Health ServicePublication 994).Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Brunner, D.W., J.H. Gillespie. Hagan’s Infectious Diseases of Domestic Animals. 6th ed.Ithaca: Coms<strong>to</strong>ck; 1973.Burden P. Actinomycosis [edi<strong>to</strong>rial]. J Infect 19:95–99, 1989.Cottral, G.E., ed. Manual of St<strong>and</strong>ardized Methods for Veterinary Microbiology. Ithaca:Coms<strong>to</strong>ck; 1978.Dalling, T., A. Robertson, eds. International Encyclopaedia of Veterinary Medicine.Edinburgh: Green; 1966.Georg, L.K. The agents of human actinomycosis. Cited in: Lerner, P.L. Actinomyces <strong>and</strong>Arachnia species. In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E. Bennett, eds. Principles <strong>and</strong>Practice of Infectious Diseases. 3rd ed. New York: Churchill Livings<strong>to</strong>ne, Inc.; 1990.Hardie, E.M., J.A. Barsanti. Treatment of canine actinomycosis. J Am Vet Assoc180:537–541, 1982.


6 BACTERIOSESKoebler, C., A. Chatwani, R. Schwartz. Actinomycosis infection associated with intrauterinecontraceptive devices. Am J Obstet Gynecol 145:596–599, 1983.Lerner, P.L. Actinomyces <strong>and</strong> Arachnia species. In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E.Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: ChurchillLivings<strong>to</strong>ne, Inc.; 1990.Pier, A.C. The actinomycetes. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger,eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.Valicenti, J.F., Jr., A.A. Pappas, C.D. Graber, H.O. Williamson, N.F. Willis. Detection <strong>and</strong>prevalence of IUD-associated Actinomyces colonization <strong>and</strong> related morbidity. A prospectivestudy of 69,925 cervical smears. JAMA 247:1149–1152, 1982.Wilson, G. Actinomycosis, actinobacillosis, <strong>and</strong> related <strong>diseases</strong>. In: Smith, G.R., ed. Vol3: Topley <strong>and</strong> Wilson’s Principles of Bacteriology, Virology <strong>and</strong> Immunity. Baltimore:Williams & Wilkins; 1984.AEROMONIASISICD-10 AO5.8 other specified bacterial foodborne in<strong>to</strong>xicationsEtiology: The genus Aeromonas is classified within the family Vibrionaceae <strong>and</strong>shares some characteristics with members of other genera of this family. However,genetic hybridization studies indicate that the genus Aeromonas is sufficiently different<strong>to</strong> place it in a new family, with the suggested name of Aeromonadaceae. Twogroups can be distinguished in the genus Aeromonas. The first group is psychrophilic<strong>and</strong> nonmotile <strong>and</strong> is represented by Aeromonas salmonicida, an importantpathogen for fish (the agent of furunculosis). It does not affect man because itcannot reproduce at a temperature of 37°C. The second group is mesophilic <strong>and</strong>motile, <strong>and</strong> it is this group that causes aeromoniasis, a disease <strong>common</strong> <strong>to</strong> man <strong>and</strong>animals. These aeromonas are gram-negative, straight bacilli ranging from 1 <strong>to</strong> 3microns in length. They have a polar flagellum <strong>and</strong> are oxidase positive <strong>and</strong> facultativelyanaerobic. They essentially include the species A. hydrophila, A. sobria, <strong>and</strong>A. caviae (J<strong>and</strong>a <strong>and</strong> Duffey, 1988), <strong>to</strong> which A. veronii <strong>and</strong> A. schuberti were addedlater, as well as the genospecies A. j<strong>and</strong>ae <strong>and</strong> A. trota. However, only A. hydrophila<strong>and</strong> A. sobria are of clinical interest.More recent hybridization studies show that the A. hydrophila complex is geneticallyvery variable. Thirteen different genospecies have been established, but froma practical st<strong>and</strong>point the three principal phenospecies are retained. It is possible <strong>to</strong>identify 95% of isolates on the basis of their biochemical properties (J<strong>and</strong>a, 1991).A system of 40 serogroups was established based on the somatic antigens (O) ofA. hydrophila <strong>and</strong> A. caviae. All the O antisera contain antibodies <strong>to</strong> the rugose form(R) of the bacillus, <strong>and</strong> thus the antisera must be absorbed by culturing the R formbefore being used (Sakazaki <strong>and</strong> Shimada, 1984). Typing is done by gel proteinelectrophoresis, isoenzyme analysis, <strong>and</strong> genetic analysis. Isoenzyme analysis madeit possible <strong>to</strong> identify genospecies through four enzymes. All these methods have


AEROMONIASIS 7shown that the clinical strains are very diverse <strong>and</strong> that no single clone is responsiblefor most of the infections (Von Graevenitz <strong>and</strong> Altwegg, 1991).Over the last decade, researchers have tried <strong>to</strong> define the virulence fac<strong>to</strong>rs of thisgenus, both in terms of structural characteristics <strong>and</strong> the extracellular products theysecrete. Considered important among the structural characteristics is a type of pilus,the “flexible” or curvilinear pilus. It is expressed when stimulated by certain environmentalconditions that give the bacteria the ability <strong>to</strong> colonize. Another structuralcharacteristic that was first discovered in au<strong>to</strong>agglutinating strains of A. salmonicidais the S layer, which is outside the cell wall. The loss of this layer—which can beseen with an electron microscope—decreases pathogenicity for fish 1,000 <strong>to</strong> 10,000times. A similar layer was later discovered in certain strains of A. hydrophila <strong>and</strong> A.sobria in infected fish <strong>and</strong> mammals, but their functional role seems <strong>to</strong> differ substantiallyfrom the same S layer in A. salmonicida (it does not make the surface ofthe bacteria hydrophobic).The substances externally secreted by aeromonas include beta-hemolysin that isproduced by certain strains of A. hydrophila <strong>and</strong> A. sobria. It has been determinedthat this hemolysin has entero<strong>to</strong>xigenic effects on lactating mice <strong>and</strong> ligated ilealloops of rabbits. Purified beta-hemolysin inoculated intravenously in<strong>to</strong> mice is lethalat a dose of 0.06 µg. The cy<strong>to</strong><strong>to</strong>nic entero<strong>to</strong>xin that causes an accumulation of fluidin the ligated ileal loop of the rabbit, as well as other effects, has also beendescribed. Between 5% <strong>and</strong> 20% of the strains produce a <strong>to</strong>xin that cross reacts withthe cholera <strong>to</strong>xin in the ELISA test (J<strong>and</strong>a, 1991).Based on tests conducted in mice <strong>and</strong> fish (the latter are much more susceptible),it can be concluded that A. hydrophila <strong>and</strong> A. sobria are more virulent than A.caviae. In addition, there is a great difference in the virulence of the strains withineach species (J<strong>and</strong>a, 1991). These variations cannot be attributed <strong>to</strong> a single virulencefac<strong>to</strong>r. In addition, it was not possible <strong>to</strong> detect a <strong>common</strong> mechanism in thepathogenic capacity of Aeromonas spp. in humans or in animals.An enzyme (acetylcholinesterase) isolated from fish infected by A. hydrophilaproved <strong>to</strong> be highly active against the central nervous system. The <strong>to</strong>xin was lethalfor fish at a dose of 0.05 µg/g of bodyweight; no lesions were observed in the tissues.The same <strong>to</strong>xin was obtained from six different strains (Nie<strong>to</strong> et al., 1991).A comparison was made of 11 environmental strains <strong>and</strong> 9 human strains. All theenvironmental strains <strong>and</strong> four of the human strains proved <strong>to</strong> be pathogenic fortrout, at a dose of 3 x 10 7 colony forming units (CFU). Only the human strainscaused death or lesions through intramuscular inoculation of mice. The virulentstrains produced more hemolysis <strong>and</strong> cy<strong>to</strong><strong>to</strong>xins in cultures at 37°C than at 28°C(Mateos et al., 1993).Geographic Distribution: The motile aeromonas appear worldwide. Their principalreservoir is in river <strong>and</strong> estuary waters, as well as in salt water where it meetsfresh water. Population density is lower in highly saline waters <strong>and</strong> waters with limiteddissolved oxygen. It has sometimes been possible <strong>to</strong> isolate Aeromonas fromchlorinated water, including muncipal water supplies. These bacteria are more prolificin summer than in winter (Stelma, 1989).Occurrence in Man: Aeromoniasis generally occurs sporadically. There is noevidence that water or foods contaminated by Aeromonas spp. have been the sourceof outbreaks (as happens with other agents, such as enterobacteria). The only cases


8 BACTERIOSESthat suggest the possibility of outbreaks are those described in 1982 <strong>and</strong> 1983. Inlate 1982, some 472 cases of gastroenteritis associated with the consumption of rawoysters occurred in Louisiana (USA). One year later, another outbreak affectedseven people in Florida. This was also attributed <strong>to</strong> raw oysters that came fromLouisiana. Pathogenicity tests were performed on 23 of the 28 strains identified asA. hydrophila; 70% tested positive in at least one of the virulence tests (Abeyta etal., 1986). There may have been other outbreaks that were not recognized becausefood <strong>and</strong> patient s<strong>to</strong>ols were not examined for detection <strong>and</strong> identification of A.hydrophila (Stelma, 1989).Occurrence in Animals: A. hydrophila is a recognized pathogen in fish, amphibians,<strong>and</strong> reptiles. The disease may occur individually or epidemically, particularlyin fish-farming pools. The agent affects many fish species, particularly fresh waterspecies. Its economic impact varies, but can be severe (S<strong>to</strong>skopf, 1993).Aeromoniasis due <strong>to</strong> A. hydrophila also causes significant illness in colonies ofamphibians <strong>and</strong> reptiles bred for experimental purposes.The Disease in Man: For some time the aeromonas were considered opportunisticbacteria. Clinical <strong>and</strong> epidemiological information amassed in recent years seems<strong>to</strong> confirm that A. hydrophila <strong>and</strong> A. sobria are the primary human pathogens, particularlyas agents of enteritis in children.The disease appears in two forms: enteric <strong>and</strong> extraenteric. Studies on the pathogenicrole of Aeromonas spp. in gastroenteritis have been conducted in Australia, theUnited States, Engl<strong>and</strong>, Thail<strong>and</strong>, <strong>and</strong>, more recently, in Rosario, Argentina (Notarioet al., 1993). Patients with <strong>and</strong> without diarrhea have been compared, with the lattergroup consisting of patients suffering from other <strong>diseases</strong> or healthy individuals.In Argentina, 8 strains (2%) were isolated from 400 fecal samples <strong>and</strong> from a colonbiopsy in children with diarrhea, <strong>and</strong> no strains were isolated from 230 childrenwithout diarrhea. In the United States, the agent was found in 1.1% of the cases <strong>and</strong>in none of the controls (Agger et al., 1985). The tests in the other countries also isolatedA. hydrophila <strong>and</strong> A. sobria with greater frequency <strong>and</strong> in greater numbersfrom diarrheal feces than from nondiarrheal feces.Enteritis due <strong>to</strong> Aeromonas spp. occurs more frequently in summer <strong>and</strong> predominantlyin children from 6 months <strong>to</strong> 5 years of age. The clinical symp<strong>to</strong>ms includeprofuse diarrhea, slight fever, <strong>and</strong> abdominal pains; vomiting is occasionally seen inpatients under 2 years of age. Cases of gastroenteritis with blood <strong>and</strong> mucus in thefeces have also been described. The disease is generally benign in children <strong>and</strong> lastsonly a few days. Gastroenteritis is much less frequent in adults, but can occur withdiarrhea of longer duration (from 10 days <strong>to</strong> several weeks or months), weight loss,<strong>and</strong> dehydration. The predominant species are A. hydrophila <strong>and</strong> A. sobria, but A.caviae has also been implicated in some cases (J<strong>and</strong>a <strong>and</strong> Duffey, 1988).The extra-intestinal clinical form can affect different organs <strong>and</strong> tissues. One very<strong>common</strong> form of contamination is through wounds <strong>and</strong> various traumas. The woundgenerally becomes infected through contact with river water, ponds, or other waterreservoirs. The most <strong>common</strong> clinical expression is cellulitis. The patient recoverscompletely in such cases.Some 20 cases have been described of infection caused by medicinal leeches(Hirudo medicinalis) used <strong>to</strong> treat pos<strong>to</strong>perative venous congestion after grafts orreplantations. The leeches inject a very powerful anticoagulant, causing the


AEROMONIASIS 9congested area <strong>to</strong> bleed for one <strong>to</strong> two hours (or longer) <strong>and</strong> preventing loss of thegraft. Leeches may harbor A. hydrophila in their digestive tract <strong>and</strong> suckers <strong>and</strong>transmit the bacteria <strong>to</strong> the patient. These infections are usually limited <strong>to</strong> contaminationof the wound, but can cause extensive tissue loss <strong>and</strong> septicemia(Lineaweaver et al., 1992).Untreated cellulitis can become complicated by myonecrosis <strong>and</strong> require amputationof a limb. If there is bacteremia, the infection may ultimately be fatal.Septicemia occurs primarily in immunodeficient patients <strong>and</strong> rarely in immunocompetentpatients. The clinical manifestations are similar <strong>to</strong> septicemia caused byother gram-negative bacteria <strong>and</strong> consist of fever <strong>and</strong> hypotension. Mortality is highin these cases (J<strong>and</strong>a <strong>and</strong> Duffey, 1988). Other clinical forms are rare.Gastroenteritis in children is a self-limiting disease <strong>and</strong> does not require treatment,except in prolonged cases. All other forms should be treated with antibiotics,such as gentamicin, amikacin, chloramphenicol, <strong>and</strong> cyprofloxacin. All strains of A.hydrophila <strong>and</strong> A. sobria are resistant <strong>to</strong> ampicillin (Gutierrez et al., 1993), A. trotais not.The Disease in Animals: Aeromoniasis is primarily a disease that affectsfish, amphibians, <strong>and</strong> reptiles. The disease is rare in wild or domestic mammals<strong>and</strong> birds.FISH: A. hydrophila is the agent of bacterial hemorrhagic septicemia in fish. Allspecies of fresh water fish are considered susceptible <strong>to</strong> this disease. The clinicalpicture is very varied <strong>and</strong> sometimes other pathogens are isolated that can confusethe diagnosis <strong>and</strong> signs of the disease. In the very acute form of the disease, deathmay occur without warning signs. In other cases, scales are lost <strong>and</strong> localizaed hemorrhagesappear in the gills, mouth, <strong>and</strong> base of the fins. Ulcers in the skin, exophthalmia,<strong>and</strong> abdomen-distending ascites may also be found. Renal <strong>and</strong> hepaticlesions are seen in very prolonged cases (S<strong>to</strong>skopf, 1993). The disease occurs sporadicallyor in outbreaks. Mortality is variable but can be high.Intensive fish farming can create conditions that favor infection, such as overpopulation<strong>and</strong> adverse environmental fac<strong>to</strong>rs (increase in organic material <strong>and</strong> decreasein dissolved oxygen). These fac<strong>to</strong>rs reduce the resistance of the fish <strong>and</strong> favor thepathogenic action of A. hydrophila <strong>and</strong> other bacteria. Pseudomonas spp. oftenaccompanies A. hydrophila in ulcerous lesions in the skin of fish (erythrodermatitis,fin disease). In northern Greece, where great losses of carp (Cyprinus carpio)occurred in ponds due <strong>to</strong> a disease characterized primarily by cutaneous ulcers, bothA. hydrophila <strong>and</strong> various species of Pseudomonas were isolated. It was possible <strong>to</strong>reproduce the disease experimentally through subcutaneous inoculation of A.hydrophila without the simultaneous presence of other bacteria (Sioutas et al.,1991). Previously, there was an outbreak in Argentina of fin disease in young blackcatfish (Rhamdia sapo). Both A. hydrophila <strong>and</strong> Pseudomonas aeruginosa were isolatedfrom fin lesions. When the disease was reproduced experimentally, there wasnot much difference between the fish inoculated with A. hydrophila alone <strong>and</strong> thoseinoculated with both bacteria (Angelini <strong>and</strong> Seigneur, 1988).Infection of striped (grey) mullet (Mugil cephalus) by A. hydrophila results in anacute septicemic disease. The agent can be isolated from the blood of mullet withthe experimentally reproduced disease one or two days after inoculation. The diseaseis characterized by inflamma<strong>to</strong>ry <strong>and</strong> proliferative changes <strong>and</strong> later by


10 BACTERIOSESnecrotic lesions. Enteritis <strong>and</strong> hepatic necrosis are constant lesions (Solimanet al., 1989).Aeromoniasis in fish can be treated with antibiotics.AMPHIBIANS: Frogs used for experimental purposes—whether in labora<strong>to</strong>rycolonies or under natural conditions—die from a disease called “red leg” that causescutaneous ulceration <strong>and</strong> septicemia. The Louisiana frog (Rana catesbeiana) sufferedvarious epizootics in 1971 <strong>and</strong> 1972. Of 4,000 tadpoles separated from theirnatural habitat <strong>and</strong> kept under labora<strong>to</strong>ry conditions, 70% died during metamorphosis<strong>and</strong> 20% died after completing it.Of the wild frogs brought <strong>to</strong> the labora<strong>to</strong>ry, 10% became ill <strong>and</strong> died during thefirst year. The tadpoles born in the labora<strong>to</strong>ry that became ill during metamorphosisdemonstrated lassitude, edema, <strong>and</strong> hemorrhage in the tail; accumulation of bloodylymph around the leg muscles; <strong>and</strong> small ulcers on the operculum <strong>and</strong> the skin ofthe abdomen. Death occurred 24 hours after onset of the disease. The disease progressedslowly in adults; it sometimes lasted up <strong>to</strong> six months <strong>and</strong> ended in death.Sick frogs had petechial or diffuse hemorrhages on the skin of their entire bodies.The lymphatic sacks were full of a bloody serous fluid <strong>and</strong> intramuscular hemorrhageswere found on the hind legs <strong>and</strong> on the periosteum (Glorioso et al., 1974).“Red leg” disease in Xenopus leavis (a frog of African origin of the familyPipidae) was described in Cuba, the United States, Great Britain, <strong>and</strong> South Africa.In Cuba, the outbreak of the disease occurred three weeks after the frogs were transferredfrom the labora<strong>to</strong>ry (where they were kept at 22°C) <strong>to</strong> ambient temperaturein order <strong>to</strong> acclimate them. The disease lasted for about 48 days <strong>and</strong> the principalsymp<strong>to</strong>ms were lethargy, anorexia, petechiae, <strong>and</strong> edema. Au<strong>to</strong>psy revealed subcutaneousedemas, hemorrhages, <strong>and</strong> ascites. Aeromonas hydrophila was isolated from14 of the 50 frogs (Bravo Fariñas et al., 1989). According <strong>to</strong> the authors, the diseasewas unleashed by environmental changes, infrequent changes of water, <strong>and</strong> traumas,as well as other fac<strong>to</strong>rs.In Johor, Malaysia, where there is a small frog-breeding industry, an outbreakoccurred that affected 80% of the animals in a population of 10,000. The disease wascharacterized by ulcers <strong>and</strong> petechial hemorrhages on the skin <strong>and</strong> opaque corneas,but no visceral lesions. In a second outbreak, the disease followed a more chroniccourse, with symp<strong>to</strong>ms such as ascites, visceral tumefaction, <strong>and</strong> nervous disorders(Rafidah et al., 1990).The indicated treatment is antibiotics <strong>to</strong> which A. hydrophila is susceptible.REPTILES: In a variety of lizards <strong>and</strong> snakes, infection due <strong>to</strong> Aeromonas is associatedwith ulcerous s<strong>to</strong>matitis. The lesions may result in septicemia, with hemorrhages<strong>and</strong> areas with ecchymoses on the integument. The animals are anorexic <strong>and</strong>suffer deterioration in their general health. One complication is pneumonia. Atau<strong>to</strong>psy, exudates are found in the lungs <strong>and</strong> secondary air passages. The viscera <strong>and</strong>gastrointestinal tract show pronounced congestion with hemorrhagic areas.Treatment consists of removing the necrotic tissue from the mouth, followed by irrigationwith 10% hydrogen peroxide. The use of such antibiotics as chloramphenicol<strong>and</strong> gentamicin is indicated (Jacobson, 1984).OTHER ANIMALS: A case was described in Nigeria of aeromoniasis in a caracallynx (Felis caracal) at a zoo. The animal was found with profuse diarrhea, anorexia,


AEROMONIASIS 11<strong>and</strong> depression. Despite antidiarrheal treatment, it died in a month. The lesions suggestedthat the cause of death was acute septicemia. A. hydrophila was isolated fromthe animal’s internal organs (Ocholi <strong>and</strong> Spencer, 1989). Similar cases had appearedin young ferrets at a research institute in Japan. The agent isolated was identified asA. sobria (Hiruma et al., 1986). A case of polyarthritis in a 3-day-old calf wasdescribed in Australia. A. hydrophila was isolated from the synovial fluid (Love <strong>and</strong>Love, 1984). In Germany, a septicemic condition attributed <strong>to</strong> A. hydrophila hasbeen described in turkeys at 3 <strong>to</strong> 16 weeks of life, with morbidity of 10% <strong>and</strong> mortalityof 1%. Cases have also been recorded in canaries <strong>and</strong> in a <strong>to</strong>ucan sufferingfrom enteritis; A. hydrophila was isolated from the viscera. A. hydrophila was isolatedin a routine postmortem examination of 15 wild, farm, <strong>and</strong> pet birds. The isolateswere taken primarily in the cold months (Shane et al., 1984). A pure culture ofA. hydrophila was isolated from a parrot (Amazona versicolor) with bilateral conjunctivitis(García et al., 1992). In all cases, the stressful conditions that contributed<strong>to</strong> the development of the disease were emphasized.Source of Infection <strong>and</strong> Mode of Transmission: The primary reservoir of A.hydrophila <strong>and</strong> A. sobria is fresh water in rivers, ponds, <strong>and</strong> lakes. It is also foundin estuaries <strong>and</strong> in low-salinity salt water. Even treated municipal water supplies cancontain Aeromonas. In a French hospital, intestinal <strong>and</strong> extraintestinal aeromoniasisin 12 patients was attributed <strong>to</strong> the drinking water (Picard <strong>and</strong> Goullet, 1987).Due <strong>to</strong> the increased numbers of motile Aeromonas in the water supply in TheNetherl<strong>and</strong>s, health authorities established maximum indicative values for the densityof these bacteria in drinking water. These values are 20 CFU/100 ml for thedrinking water in water treatment plants <strong>and</strong> 200 CFU/100 ml for water being distributed(Van der Kooij, 1988).Motile Aeromonas have not caused outbreaks with multiple cases (Altwegg et al.,1991). It is difficult <strong>to</strong> underst<strong>and</strong> why, since the bacteria are widely distributed innature, water, animal feces, <strong>and</strong> foods of animal origin, <strong>and</strong> since they also multiplyat refrigeration temperatures.The distribution of the agents in water reaches its highest level during the warmmonths, as does the disease. The situation seems <strong>to</strong> be different in tropical countries.In India, the most frequent isolates from river water occur in late winter, decliningin summer <strong>and</strong> the monsoon season (Pathak et al., 1988). These authors believe thatfish are an independent or additional reservoir, since Aeromonas can be isolatedfrom them independent of the bacteria’s density in river water.Water contaminated by virulent strains of A. hydrophila or A. sobria is the sourceof infection for man <strong>and</strong> other animals. Domestic animals, especially cattle <strong>and</strong> pigs,eliminate in their feces a large amount of Aeromonas that are probably of aquaticorigin. There are indications that, in addition <strong>to</strong> water, other contaminated foods,such as oysters <strong>and</strong> shrimp, may be a source of infection for man. A case of enteritiscaused by eating a shrimp cocktail occurred in Switzerl<strong>and</strong> in a healthy 38-yearold.Only A. hydrophila <strong>and</strong> no other pathogen was isolated from the patient’s s<strong>to</strong>ol.The strain isolated from the shrimp was biochemically identical <strong>and</strong> had the sameribosomal DNA sequence (Altwegg et al., 1991).Enteric disease occurs in normal children <strong>and</strong> the route of infection is through themouth. In contrast, both enteric <strong>and</strong> extraintestinal aeromoniasis in individuals olderthan 5 years of age occurs in combination with other conditions, such as an under-


12 BACTERIOSESlying disease, trauma, or other stress fac<strong>to</strong>rs. Wounds become infected upon contactwith water. Medicinal leeches can infect the wound they produce with theaeromonas they harbor in their digestive tract <strong>and</strong> suckers. The most serious form ofthe disease, septicemia <strong>and</strong> its various organic complications, occurs in immunodeficientindividuals <strong>and</strong> the route of infection is usually extraintestinal.Fish, amphibians, <strong>and</strong> reptiles—especially in intensive breeding programs—areinfected through the mouth. The fac<strong>to</strong>rs that contribute <strong>to</strong> infection are stress fromoverpopulation, temperature changes, lack of hygiene, <strong>and</strong> inadequate feeding.Role of Animals in the Epidemiology of the Disease: Aeromoniasis is primarilya disease <strong>common</strong> <strong>to</strong> man <strong>and</strong> animals. Fish may act as a reservoir in addition <strong>to</strong>water. Other animals contribute <strong>to</strong> contamination of the environment with their feces.Diagnosis: Diagnosis can be obtained by isolating <strong>and</strong> identifying the species ofthe etiologic agent. As a selective medium, Rimler-Shotts agar can be used; it containscitrate, novobiocin, <strong>and</strong> sodium deoxycholate as selective agents, <strong>and</strong> lysine,ornithine, <strong>and</strong> mal<strong>to</strong>se as differential agents. Another <strong>common</strong>ly used medium isagar with ampicillin <strong>and</strong> sodium deoxycholate as selective agents <strong>and</strong> trehalose as adifferential agent (García-López et al., 1993).Control: Until more is known about the disease’s epidemiology <strong>and</strong> the fac<strong>to</strong>rsthat determine its virulence, the consumption of raw foods of animal origin shouldbe avoided.Aeromonas are sensitive <strong>to</strong> heat, <strong>and</strong> pasteurization is an effective means fordestroying them in milk.The measure introduced by health authorities in The Netherl<strong>and</strong>s of setting a maximumindicative value for the density of aeromonas in the water in water treatmentplants <strong>and</strong> in the water distribution network should be considered by other countrieswhen warranted by the number of human cases.Wounds should be cleaned <strong>and</strong> disinfected <strong>to</strong> prevent contamination.In cases of replantation surgery that require the application of medical leeches, itis recommended that the patient be given antibiotics <strong>to</strong> which A. hydrophila <strong>and</strong> A.sobria are sensitive a few days prior <strong>to</strong> surgery, so as <strong>to</strong> eliminate them from thedigestive tract of the leeches.Preventing aeromoniasis in aquatic <strong>and</strong> semi-aquatic animals in intensive breedingprograms requires avoiding overpopulation, changing the water, <strong>and</strong> maintainingproper temperature <strong>and</strong> feeding regimes. Work is being done <strong>to</strong> develop vaccinesfor fish. Tests indicate that they can provide good protection (Plumb, 1984; Lamerset al., 1985; Ruangpan et al., 1986).BibliographyAbeyta, C., C.A. Kaysner, M.A. Wekell, et al. Recovery of Aeromonas hydrophila fromoysters implicated in an outbreak of foodborne illness. J Food Protect 49:643–644, 1986.Agger, W.A., J.D. McCormick, M.J. Gurwith. Clinical <strong>and</strong> microbiological features ofAeromonas hydrophila associated diarrhea. J Clin Microbiol 21:909–913, 1985.Altwegg, M., G. Martinetti Lucchini, J. Lüthy-Hottenstein, M. Rohr-Bach. Aeromonasassociatedgastroenteritis after consumption of contaminated shrimp. Europ J Clin MicrobiolInfect Dis 10:44–45, 1991.


AEROMONIASIS 13Angelini, N.M., G.N. Seigneur. Enfermedad de las aletas de Rhamdia sapo. Aislamien<strong>to</strong> delos agentes etiológicos e infección experimental. Rev Argent Microbiol 20:37–48, 1988.Bravo Fariñas, L., R.J. Monté Boada, R. Cuellar Pérez, S.C. Dumas Valdiviezo. Aeromonashydrophila, infección en Xenopus leavis. Rev Cubana Med Trop 41:208–213, 1989.García, M.E., A. Domenech, L. Dominguez, et al. Aeromonas hydrophila in a pet parrot(Amazona versicolor). Avian Dis 36:1110–1111, 1992.García-López, M.L., A. Otero, M.C. García-Fernández, J.A. San<strong>to</strong>s. Incidencia, comportamien<strong>to</strong>y control de Aeromonas hydrophila en produc<strong>to</strong>s cárnicos y lácteos. Microbiología9:49–56, 1993.Glorioso, J.C., R.L. Amborski, G.F. Amborski, D.D. Culley. Microbiological studies onsepticemic bullfrogs (Rana catesbeiana). Am J Vet Res 35:1241–1245, 1974.Gutiérrez, J., M.C. Nogales, M.C. Aretio, E. Martín. Patrón de sensibilidad de lasAeromonas spp. produc<strong>to</strong>ras de infecciones extraintestinales. An Med Interna 10:65–67, 1993.Hiruma, M., K. Ike, T. Kume. Focal hepatic necrosis in young ferrets infected withAeromonas spp. Jpn J Vet Sci 48:159–162, 1986.Jacobson, E.R. Biology <strong>and</strong> <strong>diseases</strong> of reptiles. In: Fox, J.G., B.J. Cohen, F.M. Loew, eds.Labora<strong>to</strong>ry Animal Medicine. Orl<strong>and</strong>o: Academic Press; 1984.J<strong>and</strong>a, J.M. Recent advances in the study of taxonomy, pathogenicity, <strong>and</strong> infectious syndromesassociated with the genus Aeromonas. Clin Microbiol Rev 4:397–410, 1991.J<strong>and</strong>a, J.M., P.S. Duffey. Mesophilic aeromonads in human disease: Current taxonomy, labora<strong>to</strong>ryidentification, <strong>and</strong> infectious disease spectrum. Rev Infect Dis 10:980–997, 1988.Lamers, C.H., M.J. De Haas, W.B. Muiswinkel. Humoral response <strong>and</strong> memory formationin carp after infection of Aeromonas hydrophila bacterin. Dev Comp Immunol 9:65–75, 1985.Lineaweaver, W.C., M.K. Hill, G.M. Buncke, et al. Aeromonas hydrophila infections followinguse of medicinal leeches in replantation <strong>and</strong> flap surgery. Ann Plast Surg 29:238–244, 1992.Love, R.J., D.N. Love. Aeromonas hydrophila isolated from polyarthritis in a calf. Aust VetJ 61:65, 1984.Mateos, O., J. Anguita, G. Navarro, C. Paniagua. Influence of growth temperature on theproduction of extracellular virulence fac<strong>to</strong>rs <strong>and</strong> pathogenicity of environmental <strong>and</strong> humanstrains of Aeromonas hydrophila. J Appl Bacteriol 74:111–118, 1993.Nie<strong>to</strong>, T.P., Y. San<strong>to</strong>s, L.A. Rodríguez, A.E. Ellis. An extracellular acetylcholinesteraseproduced by Aeromonas hydrophila is a major <strong>to</strong>xin for fish. Microbiol Pathogenesis11:101–110, 1991.Notario, R., E. Careno, N. Borda, et al. Aeromonas spp. en niños con síndrome diarreicoagudo. Infect Microbiol Clin 5:85–89, 1993.Ocholi, R.A., T.H. Spencer. Isolation of Aeromonas hydrophila from a captive caracal lynx(Felis caracal). J Wildl Dis 25:122–123, 1989.Pathak, S.P., J.W. Bhattache, N. Kalra, S. Ch<strong>and</strong>ra. Seasonal distribution of Aeromonashydrophila in river water <strong>and</strong> isolation from river fish. J Appl Bacteriol 65:347–352, 1988.Picard, B., Goullet P. Epidemiological complexity of hospital aeromonas infectionsrevealed by electrophoretic typing of esterases. Epidemiol Infect 98:5–14, 1987.Plumb, J.A. Immunisation des poissons d’eau chaude contre cinq agents pathogenes impartants.Symposium sur la vaccination des poissons. Paris, Office Internationale des Epizooties(OIE), 20–22 février 1984.Rafidah, J., B.L. Ong, S. Saroja. Outbreak of “red leg”—An Aeromonas hydrophila infectionin frogs. J Vet Malaysia 2:139–142, 1990.Ruangpan, L., I. Kitao, I. Yoshida. Protective efficacy of Aeromonas hydrophila vaccines innile tilapia. Vet Immunol Immunopathol 12:345–350, 1986.Sakazaki, R., T. Shimada. O-serogrouping scheme for mesophilic Aeromonas strains. Jpn JMed Sci Biol 37:247–255, 1984.Shane, S.M., K.S. Harring<strong>to</strong>n, M.S. Montrose, R.G. Roebuck. The occurrence ofAeromonas hydrophila in avian diagnostic submissions. Avian Dis 28:804–807, 1984.


14 BACTERIOSESSioutas, S., R.W. Hoffmann, C. Pfeil-Putzien, T. Scherl. Carp erythrodermatitis (CE) due <strong>to</strong>an Aeromonas hydrophila infection. J Vet Med B 38:186–194, 1991.Soliman, M.K., M. el S. Easa, M. Faisal, et al. Motile Aeromonas infection of striped (grey)mullet, Mugil cephalus. An<strong>to</strong>nie Van Leeuwenhoek 56:323–335, 1989.Stelma, G.N. Aeromonas hydrophila. In: Doyle, M.T., ed. Foodborne Bacterial Pathogens.New York: Marcel Dekker; 1989.S<strong>to</strong>skopf, M.K. Bacterial <strong>diseases</strong> of goldfish, koi <strong>and</strong> carp. In: S<strong>to</strong>skopf, M.K., ed. FishMedicine. Philadelphia: W.B. Saunders; 1993.Van der Kooij, D. Properties of aeromonads <strong>and</strong> their occurrence <strong>and</strong> hygienic significancein drinking water. Zbl Bakteriol Mikrobiol Hyg 187B:1–17, 1988.Von Graevenitz, A., M. Altwegg. Aeromonas <strong>and</strong> Plesiomonas. In: Balows, A., K.L.Herrmann, H.D. Isenberg, H.J. Shadomy, eds. Manual of Clinical Microbiology. 5th ed.Washing<strong>to</strong>n, D.C.: American Society for Microbiology; 1991.ANIMAL ERYSIPELAS AND HUMAN ERYSIPELOIDICD-10 A26.0 cutaneous erysipeloidSynonyms: Rosenbach’s erysipeloid, erythema migrans, erysipelotrichosis, rosedisease (in swine).Etiology: The etiologic agent is Erysipelothrix rhusiopathiae (E. insidiosa), agram-positive (with uneven coloration), facultatively aerobic or anaerobic, nonmotilebacillus 0.6 <strong>to</strong> 2.5 microns long that does not produce spores. When found inthe rugose phase it tends <strong>to</strong> form filaments. It is resistant <strong>to</strong> environmental fac<strong>to</strong>rs,<strong>and</strong> survives 5 days in water <strong>and</strong> 15 days in mud (Jones, 1986). The number ofserotypes is increasing: in 1987, 23 (from 1 <strong>to</strong> 23) had been recognized, with subserotypes1a, 1b <strong>and</strong> 2a, 2b (Norrung et al., 1987), <strong>and</strong> by 1991, there were already26 serotypes (Norrung <strong>and</strong> Molin, 1991). Serotyping is important in epidemiology<strong>and</strong> immunization.A second species, E. <strong>to</strong>nsillarum, was isolated from the <strong>to</strong>nsils of apparentlyhealthy swine (Takahashi et al., 1987).The classification <strong>and</strong> nomenclature of the genus Erysipelothrix is still underinvestigation. DNA:DNA hybridization studies have shown that one group of E. rhusiopathiaeserotypes is genetically more related <strong>to</strong> this species, while another isgenetically more related <strong>to</strong> E. <strong>to</strong>nsillarum. Two serotypes, 13 <strong>and</strong> 18, possiblybelong <strong>to</strong> a new species, given their low level of hybridization with both species(Takahashi et al., 1992).Geographic Distribution: The etiologic agent is distributed on all continentsamong many species of domestic <strong>and</strong> wild mammals <strong>and</strong> birds. It has also been isolatedfrom aquatic animals, such as dolphins, American alliga<strong>to</strong>rs <strong>and</strong> crocodiles,<strong>and</strong> sea lions.


ANIMAL ERYSIPELAS AND HUMAN ERYSIPELOID 15Occurrence in Man: Human erysipeloid is for the most part an occupational diseaseaffecting workers in slaughterhouses <strong>and</strong> commercial fowl-processing plants,fishermen <strong>and</strong> fish-industry workers, <strong>and</strong> those who h<strong>and</strong>le meat (particularly pork)<strong>and</strong> seafood products. It is not a notifiable disease <strong>and</strong> little is known of its incidence.In the former Soviet Union, nearly 3,000 cases were reported between 1956<strong>and</strong> 1958 in 13 slaughterhouses in the Ukraine, <strong>and</strong> 154 cases were reported in theTula region in 1959. From 1961 <strong>to</strong> 1970, the U.S. Centers for Disease Control <strong>and</strong>Prevention confirmed the diagnosis of 15 cases in the US. A few isolated cases haveoccurred in Latin America. Some epidemic outbreaks have occurred in the formerSoviet Union, in the United States, <strong>and</strong> on the southern Baltic coast (see section onsource of infection <strong>and</strong> mode of transmission).Occurrence in Animals: The disease in swine (rose disease, swine erysipelas) isimportant in Asia, Canada, Europe, Mexico, <strong>and</strong> the United States. It has also beenseen in Brazil, Chile, Guatemala, Guyana, Jamaica, Peru, <strong>and</strong> Suriname, but theincidence is low in these countries. However, the disease seems <strong>to</strong> be increasing inimportance in Chile (Skoknic et al., 1981). Polyarthritis in sheep due <strong>to</strong> E. rhusiopathiaehas been described in many sheep-breeding areas of the world.The Disease in Man: The cutaneous form is known by the name erysipeloid <strong>to</strong>distinguish it from erysipelas caused by a hemolytic strep<strong>to</strong>coccus. The incubationperiod ranges from one <strong>to</strong> seven days. Erysipeloid localizes primarily in the h<strong>and</strong>s<strong>and</strong> fingers <strong>and</strong> consists of an erythema<strong>to</strong>us, edema<strong>to</strong>us skin lesion with violet colorationaround a wound (the inoculation point) that may be a simple abrasion.Arthritis in the finger joints occurs with some frequency. The patient experiences aburning sensation, a pulsating pain, <strong>and</strong> at times an intense pruritus.The course of the disease is usually benign <strong>and</strong> the patient recovers in two <strong>to</strong> fourweeks. If the infection becomes generalized, septicemia <strong>and</strong> endocarditis may causedeath. In the US, most cases reported in recent years have been the septicemic formgenerally associated with endocarditis (McClain, 1991). An analysis of 49 cases ofsystemic infection occurring over a 15-year period (Gorby <strong>and</strong> Peacock, 1988) foundthat E. rhusiopathiae has a peculiar tropism <strong>to</strong>ward the aortic valve. In 40% of thecases, there was a concomitant cutaneous erysipeloid lesion <strong>and</strong> fatality was 38%. Inslightly more than 40%, there was a his<strong>to</strong>ry of prior valvular disease. Only 17% had ahis<strong>to</strong>ry that could be characterized as involving a compromised immune system. Theprincipal symp<strong>to</strong>ms were fever (92%), splenomegaly (36%), <strong>and</strong> hematuria (24%).Nelson (1955) did not record any cases of endocarditis among 500 cases oferysipeloid in the US, which would indicate that the systemic disease is rather rare.The first case of endocarditis in Brazil was described by Rocha et al. (1989). Thedisease began with an erysipeloid <strong>and</strong> progressed <strong>to</strong> septicemia <strong>and</strong> endocarditis.The patient was an alcoholic with a prior his<strong>to</strong>ry of aortic insufficiency, who hadpricked himself with a fishbone.The preferred treatment is penicillin, <strong>to</strong> which E. rhusiopathiae is very sensitive.Treatment with cephalosporins can be substituted for patients who are allergic <strong>to</strong>penicillin (McClain, 1991).The Disease in Animals: Many species of domestic <strong>and</strong> wild mammals <strong>and</strong> birdsare hosts <strong>to</strong> the etiologic agent. In several animal species, E. rhusiopathiae producespathologic processes. Swine are the most affected species.


16 BACTERIOSESSWINE: Swine erysipelas is an economically important disease in many countries.In several central European countries, swine can only be raised profitably where systematicvaccination is practiced. Morbidity <strong>and</strong> mortality vary a great deal from oneregion <strong>to</strong> another, perhaps due <strong>to</strong> differences in the virulence of the etiologic agent.At present, acute forms are infrequent in western Europe <strong>and</strong> in North America.The incubation period lasts from one <strong>to</strong> seven days. There are three main clinicalforms: acute (septicemia), subacute (urticaria), <strong>and</strong> chronic (arthritis, lymphadenitis,<strong>and</strong> endocarditis). These forms may coexist in a herd or appear separately. Theacute form begins suddenly with a high fever. Some animals suffer from prostration,anorexia, <strong>and</strong> vomiting, while others continue <strong>to</strong> feed despite the high fever. In someanimals, reddish purple spots appear on the skin, particularly in the ears. There issplenomegaly <strong>and</strong> swelling of the lymph nodes. In the final phase of septicemicerysipelas, dyspnea <strong>and</strong> diarrhea are the most obvious symp<strong>to</strong>ms. The disease has arapid course <strong>and</strong> mortality is usually very high (Timoney et al., 1988). The subacuteform is characterized by urticaria, which initially appears as reddish or purple rhomboid-shapedspots on the skin. These spots are found particularly on the abdomen,the inside of the thighs, the neck, <strong>and</strong> the ears. The plaques later become necrotic,dry up, <strong>and</strong> fall off.The chronic form is characterized by arthritis. At first, the joints swell <strong>and</strong> movementis painful; later, the lesion may develop in<strong>to</strong> ankylosis. Losses from arthritisare considerable because the animals’ development <strong>and</strong> weight gain are affected <strong>and</strong>because they may be confiscated from the abat<strong>to</strong>irs. The chronic form may alsoappear as endocarditis, with progressive emaciation or sudden death. Lymphadenitisis another manifestation of the chronic form (Timoney et al., 1988; Blood <strong>and</strong>Radostits, 1989).Among the isolates of E. rhusiopathiae obtained from swine with clinicalerysipelas, serotypes 1 (subtypes 1a <strong>and</strong> 1b) <strong>and</strong> 2 predominate. Subtype 1a isusually isolated from the septicemic form, serotype 2 from the urticarial <strong>and</strong> arthriticform, <strong>and</strong> serotypes 1 <strong>and</strong> 2 from endocarditis. A study conducted in Japan typed300 isolates from swine with erysipelas. Most belonged <strong>to</strong> serotypes 1a, 1b, or 2.Serotype 1a was also isolated in 9.7% of arthritis <strong>and</strong> lymphadenitis cases. Only6.7% belonged <strong>to</strong> other serotypes: 3, 5, 6, 8, 11, 21, <strong>and</strong> N (could not be typed),isolated from the chronic form of erysipelas. These latter strains were analyzedexperimentally for their pathogenicity in swine <strong>and</strong> were found <strong>to</strong> produce theurticarial form.The strains of serotype 1a isolated from swine with arthritis or lymphadenitis producedvarious symp<strong>to</strong>ms: generalized urticaria with depression <strong>and</strong> anorexia insome animals, localized urticaria lesions in other animals, <strong>and</strong> no symp<strong>to</strong>ms in theremaining animals (Takahashi, 1987).Acute cases can be treated with simultaneous administration of penicillin <strong>and</strong>antiserum.SHEEP AND CATTLE: E. rhusiopathiae causes arthritis in lambs, usually after taildocking or sometimes as a result of an umbilical infection. The infection becomesestablished about two weeks after tail docking or birth, <strong>and</strong> the main symp<strong>to</strong>ms aredifficulty in movement <strong>and</strong> stunted growth. Recovery is slow.In Argentina, Brazil, Chile, Great Britain, <strong>and</strong> New Zeal<strong>and</strong>, a cutaneous infectioncaused by E. rhusiopathiae has been observed on the hooves of sheep a few


ANIMAL ERYSIPELAS AND HUMAN ERYSIPELOID 17Figure 1. Animal erysipelas <strong>and</strong> human erysipeloid (Erysipelothrix rhusiopathiae). Mode of transmission.Infectedanimals(boar)Environmentalcontamination(pasture, water)Ingestion,through the skinSusceptibleanimals(boar)Through the skin by h<strong>and</strong>linganimals <strong>and</strong> animal products,including fishM<strong>and</strong>ays after they have undergone a benzene hexachloride dip. The lesions consist oflaminitis <strong>and</strong> the animals experience difficulty moving. The disease lasts about twoweeks. As with human erysipeloid, the infection gains entry through small skinabrasions. It can be prevented by adding a disinfectant such as a 0.03% solution ofcupric sulfate <strong>to</strong> the dip. Serotype 1b was the most <strong>common</strong> of the isolates found inAustralia, not only in swine but in domestic <strong>and</strong> wild sheep <strong>and</strong> fowl as well.Serotypes 1a <strong>and</strong> 2 were less frequent in sheep (Eamens et al., 1988).Other forms of erysipelas in sheep are valvular endocarditis, septicemia, <strong>and</strong>pneumonia (Griffiths et al., 1991).Arthritis has been observed in calves, <strong>and</strong> the agent has been isolated from the<strong>to</strong>nsils of healthy adult cows.FOWL: A septicemic disease caused by E. rhusiopathiae occurs in many speciesof domestic <strong>and</strong> wild fowl; turkeys are the most frequently affected. Symp<strong>to</strong>msinclude general weakness, diarrhea, cyanosis, <strong>and</strong> a reddish-purple swollen comb.The disease tends <strong>to</strong> attack males in particular. Mortality can vary between 2.5% <strong>and</strong>25%. The lesions consist of large hemorrhages <strong>and</strong> petechiae of the pec<strong>to</strong>ral <strong>and</strong> legmuscles, serous membranes, intestine, <strong>and</strong> gizzard. The spleen <strong>and</strong> liver areenlarged. Symp<strong>to</strong>ms <strong>and</strong> lesions are similar in chickens, ducks, <strong>and</strong> pheasants.Source of Infection <strong>and</strong> Mode of Transmission (Figure 1): Many animalspecies harbor E. rhusiopathiae. The principal reservoir seems <strong>to</strong> be swine; the etiologicagent has been isolated from the <strong>to</strong>nsils of up <strong>to</strong> 30% of apparently healthyswine. In a study carried out in Chile, the agent was isolated from <strong>to</strong>nsil samples of53.5% of 400 swine in a slaughterhouse (Skoknic et al., 1981). E. rhusiopathiae wasisolated from 25.6% of soil samples where pigs live <strong>and</strong> from their feces (Wood <strong>and</strong>


18 BACTERIOSESHarring<strong>to</strong>n, 1978). Alkaline soil is particularly favorable <strong>to</strong> the agent’s survival. Agreat variety of serotypes may be isolated from apparently healthy swine. In experimentaltests, some serotypes prove <strong>to</strong> be highly virulent, others moderately pathogenic(producing only localized urticaria), <strong>and</strong> others avirulent (Takahashi, 1987).Fish, mollusks, <strong>and</strong> crustaceans are an important source of infection. The etiologicagent has been isolated from fish skin. In the former Soviet Union, an epidemicof erysipeloid was caused by h<strong>and</strong>ling fish brought in by several differentboats; on the Baltic coast there was another outbreak of 40 cases. In Argentina,where swine erysipelas has not been confirmed but where cases of humanerysipeloid have been described, the agent was isolated from 2 out of 9 water samplesfrom the Atlantic coast, <strong>and</strong> from 1 out of 40 samples of external integument offish (de Diego <strong>and</strong> Lavalle, 1977). Subsequently, these strains were identified asbelonging <strong>to</strong> serotypes 21 <strong>and</strong> 22.In meat <strong>and</strong> poultry processing plants, rodents can be important reservoirs <strong>and</strong>dissemina<strong>to</strong>rs of the infection. Fourteen different serotypes of E. rhusiopathiae wereisolated from 38 samples (33.9%) obtained from pork in 112 shops in Tokyo. Somesamples contained more than one serotype (Shiono et al., 1990).E. rhusiopathiae can survive a long time outside the animal organism, both in theenvironment <strong>and</strong> in animal products, which contributes <strong>to</strong> its perpetuation.Man is infected through wounds <strong>and</strong> skin abrasions, but is very resistant <strong>to</strong> otherentry routes. The infection is contracted by h<strong>and</strong>ling animals <strong>and</strong> animal products,including fish. Veterinarians have contracted the infection when they pricked themselveswhile administering the simultaneous vaccination (virulent culture <strong>and</strong>serum). This procedure is no longer in use. In Chile, a case of human endocarditiswas attributed <strong>to</strong> the ingestion of smoked fish sold on the street (Gilabert, 1968).The agent can multiply in an apparently healthy carrier under stress, <strong>and</strong> cancause disease <strong>and</strong> contaminate the environment. A pig with the acute form oferysipelas sheds an enormous amount of the bacteria in its feces, urine, saliva, <strong>and</strong>vomit, thus becoming a source of infection for the other pigs on the farm (Timoneyet al., 1988).The routes of infection are believed <strong>to</strong> be digestive <strong>and</strong> cutaneous, through abrasions<strong>and</strong> wounds. The long survival of the agent in the environment ensuresendemism in affected areas. Other animals <strong>and</strong> fowl may also contribute <strong>to</strong> maintainingthe infection or <strong>to</strong> causing outbreaks.Role of Animals in the Epidemiology of the Disease: Man is an accidental hostwho contracts the infection from sick animals, carriers, animal products, or objectscontaminated by animals.Diagnosis: Clinical diagnosis, based on the patient’s occupation <strong>and</strong> on the characteristicsof the cutaneous lesion, can be confirmed by isolation <strong>and</strong> identificationof the etiologic agent. E. rhusiopathiae can be isolated from biopsies of the lesion.The sample is cultured in trypticase soy broth <strong>and</strong> incubated at 35°C for seven days;if there is growth, the culture is repeated in blood agar. The blood of septicemicpatients can be cultured directly in blood agar (Bille <strong>and</strong> Doyle, 1991).In septicemic cases in animals, the etiologic agent can be isolated from the blood<strong>and</strong> internal organs. In cases of arthritis or skin infections, cultures are made fromlocalized lesions. Isolations from contaminated materials are accomplished throughinoculation of mice, which are very susceptible.


ANIMAL ERYSIPELAS AND HUMAN ERYSIPELOID 19Diagnosis of animal erysipelas makes use of several serologic tests, such as agglutination,growth inhibition, passive hemagglutination, <strong>and</strong> complement fixation.Given the frequency of subclinical infections <strong>and</strong> vaccination in animals, serologictests are often difficult <strong>to</strong> interpret. A comparative study of the growth inhibition test<strong>and</strong> the complement fixation test concluded that the latter is more useful for diagnosis,since it eliminates low titers caused by subclinical infection or vaccination(Bercovich et al., 1981). Another serologic method is the indirect enzyme-linkedimmunosorbent assay (ELISA), which is as sensitive as the growth inhibition test<strong>and</strong> is easier <strong>and</strong> less expensive <strong>to</strong> conduct (Kirchhoff et al., 1985).Control: In persons exposed as a result of their occupations, prevention oferysipeloid primarily involves hygiene, namely frequent h<strong>and</strong> washing with disinfectant<strong>and</strong> proper treatment of wounds. Establishments where foods of animal originare processed should control rodent populations.The control of swine erysipelas depends mostly on vaccination. There are twovaccines in use that have given good results: a bacterin adsorbed on aluminumhydroxide <strong>and</strong> a live avirulent vaccine (EVA=erysipelas vaccine avirulent).Vaccination confers immunity for five <strong>to</strong> eight months. The bacterin is first administeredbefore weaning, followed by another dose two <strong>to</strong> four weeks later. The avirulentvaccine is administered orally via drinking water. The vaccines are not entirelysatisfac<strong>to</strong>ry in preventing chronic erysipelas <strong>and</strong> it is even suspected that vaccinationmay contribute <strong>to</strong> arthritic symp<strong>to</strong>ms (Timoney et al., 1988). On the other h<strong>and</strong>,the great reduction or near elimination of the acute form in western Europe, Japan,<strong>and</strong> the US is probably due <strong>to</strong> systematic vaccination. In the case of an outbreak ofsepticemic erysipelas, it is important <strong>to</strong> destroy the carcasses immediately, disinfectthe premises, <strong>and</strong> <strong>to</strong> treat sick animals with penicillin <strong>and</strong> the rest of the herd withanti-erysipelas serum. Rotation of animals <strong>to</strong> different pastures <strong>and</strong> environmentalhygiene measures are also of great help in control.Bacterins are used on turkey-raising establishments, where the infection isendemic. A live vaccine administered orally via drinking water has yielded goodresults in tests (Bricker <strong>and</strong> Saif, 1983).BibliographyBercovich, Z., C.D. Weenk van Loon, C.W. Spek. Serological diagnosis of Erysipelothrixrhusiopathiae: A comparative study between the growth inhibition test <strong>and</strong> the complementfixation test. Vet Quart 3:19–24, 1981.Bille, J., M.P. Doyle. Listeria <strong>and</strong> Erysipelothrix. In: Ballows, A., W.J. Hausler, Jr., K.L.Hermann, et al. Manual of Clinical Microbiology. 5th ed. Washing<strong>to</strong>n, D.C.: AmericanSociety for Microbiology; 1991.Blood, D.C., O-M. Radostits. Veterinary Medicine. 7th ed. London: Baillière Tindall; 1989.Bricker, J.M., Y.M. Saif. Drinking water vaccination of turkeys, using live Erysipelothrixrhusiopathiae. J Am Vet Med Assoc 183:361–362, 1983.de Castro, A.F.P., O. Campedelli Filho, C. Troise. Isolamen<strong>to</strong> de Erysipelothrix rhusiopathiaede peixes maritimos. Rev Inst Med Trop Sao Paulo 9:169–171, 1967.de Diego, A.I., S. Lavalle. Erysipelothrix rhusiopathiae en aguas y pescados de la costaatlántica de la provincia de Buenos Aires (Argentina). Gac Vet 39:672–677, 1977.Eamens, G.J., M.J. Turner, R.E. Catt. Serotypes of Erysipelothrix rhusiopathiae inAustralian pigs, small ruminants, poultry, <strong>and</strong> captive wild birds <strong>and</strong> animals. Aust Vet J65:249–252, 1988.


20 BACTERIOSESGilabert, B. Endocarditis bacteriana producida por Erysipelothrix. Primer caso humanoverificado en Chile. Bol Hosp San Juan de Dios 15:390–392, 1968. Cited in: Skoknic, A., I.Díaz, S. Urcelay, R. Duarte, O. González. Estudio de la erisipela en Chile. Arch Med Vet13:13–16, 1981.Gledhill, A.W. Swine erysipelas. In: Stableforth, A.W., I.A. Galloway, eds. InfectiousDiseases of Animals. London: Butterworths; 1959.Gorby, G.L., J.E. Peacock, Jr. Erysipelothrix rhusiopathiae endocarditis: Microbiologic,epidemiologic, <strong>and</strong> clinical features of an occupational disease. Rev Infect Dis 10:317–325,1988.Griffiths, I.B., S.H. Done, S. Readman. Erysipelothrix pneumonia in sheep. Vet Rec128:382–383, 1991.Jones, D. Genus Erysipelothrix, Rosenbach 1909. In: Sneath, P.H., H.S. Mair, M.E. Sharpe,eds. Vol. 2: Bergey’s Manual of Systemic Bacteriology. Baltimore: Williams & Wilkins; 1986.Kirchhoff, H., H. Dubenkroop, G. Kerlen, et al. Application of the indirect enzymeimmunoassay for the detection of antibodies against Erysipelothrix rhusiopathiae. VetMicrobiol 10:549–559, 1985.Levine, N.D. Listeriosis, botulism, erysipelas, <strong>and</strong> goose influenza. In: Biester, H.E., L.H.Schwarte, eds. Diseases of Poultry. 4th ed. Ames: Iowa State University Press; 1959.McClain, J.B. Erysipelothrix rhusiopathiae In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E.Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: ChurchillLivings<strong>to</strong>ne, Inc.; 1990.Nelson, E. Five hundred cases of erysipeloid. Rocky Mt Med J 52:40–42, 1955. Cited in:Gorby, G.L., J.E. Peacock, Jr. Erysipelothrix rhusiopathiae endocarditis: Microbiologic, epidemiologic,<strong>and</strong> clinical features of an occupational disease. Rev Infect Dis 10:317–325, 1988.Norrung, V., G. Molin. A new serotype of Erysipelothrix rhusiopathiae isolated from pigslurry. Acta Vet Hung 39:137–138, 1991.Norrung, V., B. Munch, H.E. Larsen. Occurrence, isolation <strong>and</strong> serotyping of Erysipelothrixrhusiopathiae in cattle <strong>and</strong> pig slurry. Acta Vet Sc<strong>and</strong> 28:9–14, 1987.Rocha, M.P., P.R.S. Fon<strong>to</strong>ura, S.N.B. Azevedo, A.M.V. Fon<strong>to</strong>ura. Erysipelothrix endocarditiswith previous cutaneous lesion: Report of a case <strong>and</strong> review of literature. Rev Inst MedTrop Sao Paulo 31:286–289, 1989.Shiono, H., H. Hayashidani, K-I. Kaneko, et al. Occurrence of Erysipelothrix rhusiopathiaein retail raw pork. J Food Protect 53:856–858, 1990.Shuman, R.D., R.L. Wood. Swine erysipelas. In: Dunne, H.W., ed. Diseases of Swine. 3rded. Ames: Iowa State University Press; 1970.Skoknic, A., I. Díaz, S. Urcelay, R. Duarte, O. González. Estudio de la erisipela en Chile.Arch Med Vet 13:13–16, 1981.Takahashi, T. Studies on serotypes, antibiotic resistance, <strong>and</strong> pathogenic characteristics ofErysipelothrix rhusiopathiae. Bull Nipon Vet Zootechn College 36:153–156, 1987.Takahashi, T., T. Fujisawa, T. Benno, et al. Erysipelothrix <strong>to</strong>nsillarum sp. nov. isolated from<strong>to</strong>nsils of apparently healthy pigs. Int J Syst Bacteriol 37:166–169, 1987.Takahashi, T., T. Fujisawa, Y. Tamura, et al. DNA relatedness among Erysipelothrix rhusiopathiaestrains representing all twenty-three serovars <strong>and</strong> Erysipelothrix <strong>to</strong>nsillarum. Int JSyst Bacteriol 42:469–473, 1992.Timoney, J.F., J.H. Gillespie, F.W. Scott, J.E. Barlough. Hagan <strong>and</strong> Bruner’s Microbiology<strong>and</strong> Infectious Diseases of Domestic Animals. 8th ed. Ithaca: Coms<strong>to</strong>ck; 1988.Wood, R.L. Erysipelothrix infection. In: Hubbert, W.T., W.F. McCulloch, P.R.Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield:Thomas; 1975.Wood, R.L., R. Harring<strong>to</strong>n. Serotypes of Erysipelothrix rhusiopathiae isolated fromswine <strong>and</strong> from soil <strong>and</strong> manure of swine pens in the United States. Am J Vet Res39:1833–1840, 1978.


ANTHRAX 21Wood, R.L., R. Harring<strong>to</strong>n, D.R. Hubrich. Serotypes of previously unclassified isolates ofErysipelothrix rhusiopathiae from swine in the United States <strong>and</strong> Puer<strong>to</strong> Rico. Am J Vet Res42:1248–1250, 1981.ANTHRAXICD-10 A22.0 cutaneous anthrax; A22.1 pulmonary anthrax;A22.2 gastrointestinal anthraxSynonyms: Malignant pustule, malignant carbuncle, charbon, hematic anthrax,bacterial anthrax, splenic fever, woolsorters’ disease.Etiology: Bacillus anthracis, an aerobic, nonmotile, gram-positive bacillus 3–5microns long that forms centrally located spores. It should be differentiated from B.cereus, which is quite similar. One of the media used <strong>to</strong> differentiate them is thegamma phage specific for B. anthracis. The etiologic agent is found in a vegetativestate in man <strong>and</strong> animals. When exposed <strong>to</strong> oxygen in the air, it forms spores thatare highly resistant <strong>to</strong> physical <strong>and</strong> chemical agents.In nature, B. anthracis occurs in a virulent form—the pathogenic agent ofanthrax—<strong>and</strong> in an avirulent form. Virulence is determined by a capsule that inhibitsphagocy<strong>to</strong>sis <strong>and</strong> an exo<strong>to</strong>xin, both of which are plasmid mediated. In turn, the <strong>to</strong>xinconsists of three protein fac<strong>to</strong>rs: the protective antigen, the lethal fac<strong>to</strong>r, <strong>and</strong> theedema fac<strong>to</strong>r. None of these fac<strong>to</strong>rs is <strong>to</strong>xic by itself. When injected intravenouslyat the same time, the protective antigen <strong>and</strong> the lethal fac<strong>to</strong>r are lethal in some animalspecies. The combination of the protective agent <strong>and</strong> the edema fac<strong>to</strong>r producesedema when injected subcutaneously (Little <strong>and</strong> Knudson, 1986).Geographic Distribution: Worldwide, with areas of enzootic <strong>and</strong> sporadicoccurrence.Occurrence in Man: The infection in humans is correlated with the incidence ofthe disease in domestic animals. In economically advanced countries, where animalanthrax has been controlled, it occurs only occasionally among humans. Some casesstem from the importation of contaminated animal products. Human anthrax is most<strong>common</strong> in enzootic areas in developing countries, among people who work withlives<strong>to</strong>ck, eat undercooked meat from infected animals, or work in establishmentswhere wool, goatskins, <strong>and</strong> pelts are s<strong>to</strong>red <strong>and</strong> processed. The incidence of humanillness in developing countries is not well known because those sick with the diseasedo not always see a doc<strong>to</strong>r, nor do doc<strong>to</strong>rs always report the cases; in addition, thediagnosis often is based only on the clinical syndrome.According <strong>to</strong> data from recent years, epidemic outbreaks continue <strong>to</strong> occurdespite the availability of excellent preventive measures for animal anthrax <strong>and</strong>,therefore, for the occurrence of the disease in humans. There are some hyperendemicareas, as was shown in Haiti when an American woman contracted the infec-


22 BACTERIOSEStion after acquiring some goatskin drums. Compilation of data in that countryrevealed a high incidence of human anthrax in the southern peninsula, Les Cayes,which has a population of approximately 500,000. From 1973 <strong>to</strong> 1977, 1,587 caseswere recorded in the 31 clinics in that region (La Force, 1978).In Zambia, at least 30 people died from anthrax in 1992. Eastern Nigeria has avery high incidence of human anthrax (Okolo, 1985). On the borders betweenThail<strong>and</strong>, Myanmar (Burma), <strong>and</strong> Laos that are crossed by animals transported fromas far away as India, outbreaks occur frequently. In one Thai village, several of theapproximately 200 inhabitants participated in cutting up a buffalo that had supposedlydrowned; eight of them became ill <strong>and</strong> one died with symp<strong>to</strong>ms suspected ofbeing anthrax (Ngampachjana et al., 1989). In a settlement in eastern Algeria, 6cases of anthrax occurred in an extended family of 59 members. Those who fell illhad participated in slaughtering <strong>and</strong> butchering a sheep with symp<strong>to</strong>ms thatincluded hemorrhage, black blood, <strong>and</strong> splenomegaly. Fourteen animals of variousruminant species had died before the appearance of the index case, a child who laterdied (Abdenour et al., 1987). In the former Soviet Union, at least 15,000 cases ofhuman anthrax occurred prior <strong>to</strong> 1917 <strong>and</strong> 178 cases were reported as late as 1985(Marshall, 1988).In enzootic areas, the human disease is usually endemosporadic with epidemicoutbreaks. The latter are caused primarily by ingestion of meat, often by many people,from animals who were dead or dying from anthrax when slaughtered (Rey etal., 1982; Fragoso <strong>and</strong> Villicaña, 1984; Sirisanthana et al., 1984). In 1978, in aregion in the Republic of Mali, there were 84 cases with 19 deaths. High mortality,possibly due <strong>to</strong> intestinal anthrax, was also seen in Senegal in 1957, with 237 deathsout of 254 cases (Simaga et al., 1980).In 1979, an epidemic outbreak in Sverdlovsk, in the former Soviet Union, led <strong>to</strong>a controversy between that country <strong>and</strong> the United States. According <strong>to</strong> the formerSoviet Union, fewer than 40 people died from gastric anthrax in this epidemic, whileUS intelligence sources claimed that several hundred <strong>to</strong> a thous<strong>and</strong> people perishedfrom pulmonary anthrax within a few weeks. Later Soviet sources indicated a <strong>to</strong>talof 96 victims, 79 suffering from intestinal infection (64 of whom died), <strong>and</strong> no pulmonarycases (Marshall, 1988). The controversy centered on whether the epidemicwas natural or man-induced, since the US intelligence source suspected that an accidenthad occurred at a plant presumably engaged in biological warfare projects. Ifso, this would have indicated a violation of the 1975 treaty against biologicalweapons (Wade, 1980). Sverdlovsk is located in an enzootic area <strong>and</strong>, according <strong>to</strong>Marshall (1988), the source of infection was probably a bone meal food supplemen<strong>to</strong>n State-run <strong>and</strong> private farms. Using preserved tissue, Russian <strong>and</strong> Americanresearchers were ultimately able <strong>to</strong> determine that at least 42 people had died frominhaling rather than ingesting the etiological agent. They thus confirmed the suspicionthat the source of infection was airborne <strong>and</strong> probably came from an illegalplant that the Soviet authorities did not allow <strong>to</strong> be inspected.Occurrence in Animals: Anthrax is <strong>common</strong> in enzootic areas where no controlprograms have been established.In a hyperenzootic area of eastern Nigeria, animals submitted for emergencyslaughter were studied. There is no ante mortem inspection of animals in that region,thus increasing the risk of human exposure. Of 150 animals, 34 (22.7%) were posi-


ANTHRAX 23tive through culture <strong>and</strong> inoculation of labora<strong>to</strong>ry animals. Of 35 cows, 42.9%were positive, <strong>and</strong> of 70 bulls, 14.3% were positive. The milk from 43 cows <strong>and</strong> 8sheep was also examined; 15 <strong>and</strong> 2 of the samples, respectively, were positive(Okolo, 1988).Some outbreaks <strong>and</strong> occasional cases of human infection have also been reportedin industrialized countries, such as the US (Hunter et al., 1989).In Africa, wildlife reserves periodically suffer great losses, especially among herbivores.A thesis presented at the University of Nairobi, Kenya, estimated thatanthrax accounts for about 11% of the mortality in the animal population each year,excluding calves. At E<strong>to</strong>sha National Park in Namibia, anthrax caused the death of1,635 wild animals of 10 species, or 54% of <strong>to</strong>tal mortality between January 1966<strong>and</strong> June 1974. The source of infection was artificial ponds (Ebedes, 1976). An outbreakoccurred on a reserve in Zambia between June <strong>and</strong> November of 1987, with a<strong>to</strong>tal loss of over 4,000 animals. The victims were primarily hippopotamuses(Hippopotamus amphibius). Other species, such as the Cape buffalo (Syncerus caffer)<strong>and</strong> the elephant (Loxodonta africana), also seem <strong>to</strong> have been affected(Turnbull et al., 1991).The Disease in Man: The incubation period is from two <strong>to</strong> five days. Three clinicalforms are recognized: cutaneous, pulmonary or respira<strong>to</strong>ry, <strong>and</strong> gastrointestinal.The cutaneous form is the most <strong>common</strong> <strong>and</strong> is contracted by contact withinfected animals (usually carcasses) or contaminated wool, hides, <strong>and</strong> fur. Theexposed part of the skin begins <strong>to</strong> itch <strong>and</strong> a papule appears at the inoculation site.This papule becomes a vesicle <strong>and</strong> then evolves in<strong>to</strong> a depressed, black eschar.Generally, the cutaneous lesion is not painful or is only slightly so; consequently,some patients do not consult a doc<strong>to</strong>r in time. If left untreated, the infection can lead<strong>to</strong> septicemia <strong>and</strong> death. The case fatality rate for untreated anthrax is estimated atbetween 5% <strong>to</strong> 20%.The pulmonary form is contracted by inhalation of B. anthracis spores. Atthe onset of illness, the symp<strong>to</strong>ma<strong>to</strong>logy is mild <strong>and</strong> resembles that of a <strong>common</strong>upper respira<strong>to</strong>ry tract infection. Thus, many patients do not see a doc<strong>to</strong>r in the earlystage of the disease when it would be easily cured. Some three <strong>to</strong> five days later thesymp<strong>to</strong>ms become acute, with fever, shock, <strong>and</strong> resultant death. The case fatalityrate is high.Gastrointestinal anthrax is contracted by ingesting meat from infected animals<strong>and</strong> is manifested by violent gastroenteritis with vomiting <strong>and</strong> bloody s<strong>to</strong>ols.Mortality ranges from 25% <strong>to</strong> 75% (Brachman, 1984).The recommended treatment for cutaneous anthrax is intramuscular administrationof 1 million units of procaine penicillin every 12 <strong>to</strong> 24 hours for five <strong>to</strong> sevendays. In the case of serious illness, as in pulmonary anthrax, the recommendation is2 million units of penicillin G per day administered intravenously or 500,000 unitsadministered intravenously through a slow drip every four <strong>to</strong> six hours until temperaturereturns <strong>to</strong> normal. Strep<strong>to</strong>mycin, in 1 g <strong>to</strong> 2 g doses per day, has a synergisticeffect if administered at the same time as penicillin. Some penicillin-resistantstrains of B. anthracis have been found (Braderic <strong>and</strong> Punda-Polic, 1992). Penicillinsterilizes the organism in a short time, even in a single day in patients suffering fromcutaneous anthrax, but it should be borne in mind that the <strong>to</strong>xin remains <strong>and</strong> thepatient is still not cured.


24 BACTERIOSESThe Disease in Animals: It takes three forms: apoplectic or peracute, acute <strong>and</strong>subacute, <strong>and</strong> chronic. The apoplectic form is seen mostly in cattle, sheep, <strong>and</strong>goats, <strong>and</strong> occurs most frequently at the beginning of an outbreak. The onset is sudden<strong>and</strong> death ensues rapidly. The animals show signs of cerebral apoplexy <strong>and</strong> die.The acute <strong>and</strong> subacute forms are frequent in cattle, horses, <strong>and</strong> sheep. The symp<strong>to</strong>ma<strong>to</strong>logyconsists of fever, a halt <strong>to</strong> rumination, excitement followed by depression,respira<strong>to</strong>ry difficulty, uncoordinated movements, convulsions, <strong>and</strong> death.Bloody discharges from natural orifices as well as edemas in different parts of thebody are sometimes observed.Chronic anthrax occurs mainly in less susceptible species, such as pigs, but is alsoseen in cattle, horses, <strong>and</strong> dogs. During outbreaks in swine herds, some animals fallvictim <strong>to</strong> the acute form, but most suffer from chronic anthrax. The main symp<strong>to</strong>mof this form is pharyngeal <strong>and</strong> lingual edema. Frequently, a foamy, sanguinolent dischargefrom the mouth is observed. The animals die from asphyxiation. Anotherlocalized chronic form in pigs is intestinal anthrax.Anthrax also affects free-roaming wild animals <strong>and</strong> those in zoos <strong>and</strong> nationalparks (see section on occurrence in animals).Au<strong>to</strong>psies of acute cases reveal bloody exudate in the natural orifices.Decomposition is rapid <strong>and</strong> the carcass becomes bloated with gases. Rigor mortis isincomplete. Hemorrhages are found in the internal organs; splenomegaly is almostalways present (but may not be in some cases), with the pulp being dark red or blackish<strong>and</strong> having a soft or semifluid consistency; the liver, kidneys, <strong>and</strong> lymph nodesare congested <strong>and</strong> enlarged; <strong>and</strong> the blood is blackish with little clotting tendency.Animals treated early with penicillin recover. Treatment consists of intravenousadministration of 12,000 <strong>to</strong> 17,000 units/kg of bodyweight of sodium benzylpenicillinfollowed by intramuscular administration of amoxicillin.Source of Infection <strong>and</strong> Mode of Transmission (Figure 2): Soil is the reservoirfor the infectious agent. The process followed by spores in the earth is a subject ofcontroversy. It has been suggested that there is a cycle of germination <strong>and</strong> subsequentresporulation, but there is no evidence <strong>to</strong> this effect. The lifecycle of spores under labora<strong>to</strong>ryconditions (in culture media) or in sterile soil is extremely long. However,under natural conditions, it seems that their survival is limited <strong>to</strong> a few years, due <strong>to</strong>the activity of saprophytic microbes in the soil. This is probably the case in wild animalreserves in Africa, where attempts <strong>to</strong> isolate B. anthracis from the soil or waterone or two years after an epizootic yielded negative results, except near the remainsof some animals that died from sporadic cases of anthrax. Turnbull et al. (1991)believe that in order for an enzootic area <strong>to</strong> be maintained, it would be necessary forthe etiologic agent <strong>to</strong> multiply in animals. However, a fact <strong>to</strong> be noted is the long survivalof B. anthracis on the Scottish isl<strong>and</strong> of Gruinard, which was abundantly seededwith B. anthracis during the Second World War for purposes of experimentation withbiological weapons. Some 40 years later, viable spores of B. anthracis were stilldetected. It is speculated that this long survival is due <strong>to</strong> the isl<strong>and</strong>’s acidic soil <strong>and</strong>cold, moist climate, which are unfavorable <strong>to</strong> the activity of microbial flora.For man, the source of infection is always infected animals, contaminated animalproducts, or environmental contamination by spores from these sources.Cutaneous anthrax is contracted by inoculation during the process of skinning orbutchering an animal or by contact with infected leather, pelts, wool, or fur. Broken


ANTHRAX 25Figure 2. Anthrax. Transmission cycle.Cows <strong>and</strong>calves, sheep,goats, horses,swine, dogsinfected withB. anthracisGround contaminatedwith open cadavers,secretions, <strong>and</strong>excretionsIngestionSusceptible(not vaccinated)cows <strong>and</strong> calves,sheep, goats,horses, swine,dogsMainly by contact with dead animals,hides, wool; ingestion of anthracicmeat; inhalation of sporesManskin favors transmission. Products made from contaminated hair (e.g., shavingbrushes), skins (e.g., drums), <strong>and</strong> bone meal (e.g., fertilizer) may continue <strong>to</strong> besources of infection for many years. Transmission from animals <strong>to</strong> man is possibleby means of insects acting as mechanical vec<strong>to</strong>rs, but reliably documented cases arefew. A recent case occurred in a Croatian villager who was probably stung by ahorsefly <strong>and</strong> developed a case of cutaneous anthrax on the back of her neck. The presumedsource of infection was a cow close <strong>to</strong> her home that had died of anthrax(Braderic <strong>and</strong> Punda-Polic, 1992).Pulmonary anthrax comes from inhaling spores released from contaminated woolor animal hair.The source of infection for the gastrointestinal form is domestic <strong>and</strong> wild animalsthat died from anthrax. The pathway of transmission is through the digestive tract.Cases have been observed in Asia, Africa, <strong>and</strong> Latin America.Animals contract the infection mainly by ingestion of pasture or water contaminatedwith B. anthracis spores, especially in places near anthrax-infected carcasses.An animal dying of anthrax produces an enormous quantity of B. anthracis in its tissues,<strong>and</strong> if the carcass is opened, the bacilli sporulate, contaminating the soil, grass,<strong>and</strong> water. Animals that graze in the contaminated area become infected themselves<strong>and</strong> produce new foci of infection. Animals <strong>and</strong> birds that feed on carrion can transportthe infection some distance. The most serious outbreaks occur during dry summersafter heavy rains. The rain washes spores loose <strong>and</strong> concentrates them in lowspots, forming so-called “cursed fields,” that are usually damp areas with glacialcalcareous soils containing abundant organic material <strong>and</strong> having a pH above 6 (VanNess, 1971). Nevertheless, outbreaks of anthrax may occur in acidic soil, as hap-


26 BACTERIOSESpened in the 1974 epizootic in Texas (USA), during which 218 cattle, 6 horses, <strong>and</strong>1 mule died. Eighty-three percent of the fields where the outbreak <strong>to</strong>ok place hadacid pH soil <strong>and</strong> 94% had subsoil with an alkaline pH (Whitford, 1979).Contaminated animal by-products, especially bone meal <strong>and</strong> blood meal used asfood supplements, can also give rise <strong>to</strong> distant foci of infection.Another mode of transmission is cutaneous entry through insect bites, but this isconsidered of minor epidemiologic importance.Role of Animals in the Epidemiology of the Disease: Animals are essential.Anthrax is transmitted <strong>to</strong> humans by animals or animal products. Transmissionbetween humans is exceptional.Diagnosis: The presence of the etiologic agent must be confirmed by microscopicexamination of stained smears of vesicular fluid (in man), edemas (in swine), <strong>and</strong>blood (in other animals); by culturing the microorganism from the liquid aspiratedfrom malignant pustules or from blood samples of a dead or dying animal; <strong>and</strong> byinoculation of labora<strong>to</strong>ry animals (guinea pigs <strong>and</strong> mice). If the material is contaminated,cutaneous inoculation (by scarification) should be used. The use of antibioticsquickly reduces the possibility of isolating the etiologic agent.The fluorescent antibody technique applied <strong>to</strong> fresh stains or blood smears canprove useful for presumptive diagnosis. Smears of blood or other bodily fluids canalso be stained using the Giemsa or Wright method <strong>to</strong> make the pink capsule thatsurrounds the bacillus st<strong>and</strong> out. The Ascoli precipitation test has limited value due<strong>to</strong> its limited specificity, but is still used in some labora<strong>to</strong>ries for animal products,from which the agent cannot be isolated.The ELISA <strong>and</strong> Western Blot tests can be used <strong>to</strong> detect antibodies <strong>to</strong> the protectiveantigen in individuals who have had anthrax <strong>and</strong> from whom the agent cannotbe isolated, i.e., in retrospective studies (Thurnbull et al., 1986; Sirisanthana et al.,1988; Harrison et al., 1989). Antibodies have also been found in people living nearanimal reserves in Africa who have been exposed <strong>to</strong> anthrax in wild animals withoutbecoming ill themselves (Thurnbull et al., 1991).Control: In man, the prevention of anthrax is based mainly on: (a) control of theinfection in animals; (b) prevention of contact with infected animals <strong>and</strong> contaminatedanimal products; (c) environmental <strong>and</strong> personal hygiene in places whereproducts of animal origin are h<strong>and</strong>led (adequate ventilation <strong>and</strong> work clothing); (d)medical care for cutaneous lesions; <strong>and</strong> (e) disinfection of fur <strong>and</strong> wool with hotformaldehyde. Occupational groups at risk may benefit from vaccination with theprotective antigen.The human vaccine used in the US <strong>and</strong> Great Britain is acellular <strong>and</strong> consists ofa filtrate of B. anthracis culture from a nonencapsulated strain that is adsorbed withaluminum hydroxide. This vaccine is not very potent <strong>and</strong> may not protect against allfield strains. In the countries of Eastern Europe <strong>and</strong> in China, a live attenuated sporevaccine is administered by scarification.In animals, anthrax control is based on systematic vaccination in enzootic areas.Sterne’s avirulent spore vaccine is indicated because of its effectiveness <strong>and</strong> safety.The vaccine consists of spores from the nonencapsulated 34F2 strain with an adjuvant—usuallya saponin—<strong>and</strong> is currently used worldwide, with a few exceptions.It is suitable for all domestic animal species. However, goats sometimes have severe


ANTHRAX 27reactions <strong>and</strong> the recommendation is thus <strong>to</strong> administer the vaccine in two doses inthis species, with a month between doses (administer one-fourth of the dose in thefirst month <strong>and</strong> the full dose the following month). Pregnant females of any speciesshould not be vaccinated unless they are at high risk of contracting anthrax.Antibiotics should not be administered a few days before or a few days after vaccination.In general, annual vaccination is sufficient; only in hyperenzootic areas isvaccination at shorter intervals recommended. Immunity is established in approximatelyone week in cattle, but takes longer in horses. In regions where anthraxoccurs sporadically, mass vaccination is not justified <strong>and</strong> should be limited <strong>to</strong>affected herds. Rapid diagnosis, isolation, <strong>and</strong> treatment of sick animals with antibiotics(penicillin) are important.Au<strong>to</strong>psies should not be performed on animals that have died from anthrax. Anunopened carcass decomposes rapidly <strong>and</strong> the vegetative form of B. anthracis isdestroyed in a short time. To make the diagnosis, it is recommended that blood betaken from a peripheral vessel with a syringe <strong>and</strong> sent <strong>to</strong> the labora<strong>to</strong>ry in a sterilecontainer. Dead animals should be destroyed where they lie as quickly as possible,preferably by incineration. The alternative is <strong>to</strong> bury them two meters deep <strong>and</strong>cover them with a layer of lime.In areas where these procedures are not possible, the dead animal should be leftintact so that it will start <strong>to</strong> decompose <strong>and</strong>, as much as possible, natural orifices <strong>and</strong>the surrounding soil should be treated with 10% formol (25% formalin).Affected herds should be placed in quarantine, which should last until two weeksafter the last case is confirmed, with no animal or animal product allowed out.If anthrax is suspected at a slaughterhouse, all operations should be halted untilthe diagnosis is confirmed. If positive, all exposed carcasses should be destroyed<strong>and</strong> the premises carefully disinfected (with a 5% caustic lye solution for eighthours) before operations are resumed.BibliographyAbdenour D., B. Larouze, D. Dalichaouche, M. Aouati. Familial occurrence of anthrax inEastern Algeria [letter]. J Infect Dis 155:1083–1084, 1987.Brachman, P.S. Anthrax. In: Warren, K.S., A.A.F. Mahmoud, eds. Tropical <strong>and</strong>Geographical Medicine. New York: McGraw-Hill Book Co.; 1984.Braderic N., V. Punda-Polic. Cutaneous anthrax due <strong>to</strong> penicillin-resistant Bacillusanthracis transmitted by an insect bite [letter]. Lancet 340:306–307, 1992.Ebedes, H. Anthrax epizootics in wildlife in the E<strong>to</strong>sha Park, South West Africa. In: Page,L.A., ed. Wildlife Diseases. New York: Plenum Press; 1976.Fragoso Uribe, R., H. Villicaña Fuentes. Antrax en dos comunidades de Zacatecas, México.Bol Oficina Sanit Panam 97:526–533, 1984.Harrison, L.H, J.W. Ezzel, T.G. Abshire, et al. Evaluation of serologic tests for diagnosis ofanthrax after an outbreak of cutaneous anthrax in Paraguay. J Infect Dis 160:706–710, 1989.Hunter, L., W. Corbett, C. Grinden. Anthrax. Zoonoses update. J Am Vet Med Assoc194:1028–1031, 1989.La Force, F.M. Informe a la Oficina Sanitaria Panamericana. Haiti, 1978.Little, S.F., G.B. Knudson. Comparative efficacy of Bacillus anthracis live spore vaccine<strong>and</strong> protective antigen vaccine against anthrax in the guinea pig. Infect Immun 52:509–512, 1986.


28 BACTERIOSESMarshall, E. Sverdlovsk: Anthrax capital? [news <strong>and</strong> comments]. Science 240:383–385, 1988.Ngampochjana, M., W.B. Baze, A.L Chedester. Human anthrax [letter]. J Am Vet MedAssoc 195:167, 1989.Okolo, M.I. Studies on anthrax in food animals <strong>and</strong> persons occupationally exposed <strong>to</strong> the<strong>zoonoses</strong> in Eastern Nigeria. Int J Zoonoses 12:276–282, 1985.Okolo, M.I. Prevalence of anthrax in emergency slaughtered food animals in Nigeria. VetRec 122:636, 1988.World Health Organization. Joint FAO/<strong>WHO</strong> Expert Committee on Zoonoses. Third Report.Geneva: <strong>WHO</strong>; 1967. (Technical Report Series 378).Rey, J.L., M. Meyran, P. Saliou. Situation épidémiologique de charbon human en HauteVolta. Bull Soc Pathol Exot 75:249–257, 1982.Simaga, S.Y., E. As<strong>to</strong>rquiza, M. Thiero, R. Baylet. Un foyer de charbon humain et animaldans le cercle de Kati (République du Mali). Bull Soc Pathol Exot 73:23–28, 1980.Sirisanthana, T., M. Navachareon, P. Tharavichitkul, et al. Outbreak of oral-pharyngealanthrax: An unusual manifestation of human infection with Bacillus anthracis. Am J Trop MedHyg 33:144–150, 1984.Sirisanthana, T., K.E. Nelson, J.W. Ezzell, T.G. Abshire. Serological studies of patients withcutaneous <strong>and</strong> oral-orophangyngeal anthrax from northern Thail<strong>and</strong>. Am J Trop Med Hyg39:575–581, 1988.Sirol, J.,Y. Gendron, M. Condat. Le charbon humain en Afrique. Bull <strong>WHO</strong> 49:143–148, 1973.Sterne, M. Anthrax. In: Stableforth, A.W., I.A. Galloway, eds. Infectious Diseases ofAnimals. London: Butterworths; 1959.Turnbull, P.C., R.H. Bell, K. Saigawa, et al. Anthrax in wildlife in the Luangwa Valley,Zambia. Vet Rec 128:399–403, 1991.Turnbull, P.C., M.G. Broster, A. Carman, et al. Development of antibodies <strong>to</strong> protectiveantigen <strong>and</strong> lethal fac<strong>to</strong>r components of anthrax <strong>to</strong>xin in humans <strong>and</strong> guinea pigs <strong>and</strong> their relevance<strong>to</strong> protective immunity. Infect Immun 52:356–363, 1986.Van Ness, G.B. Ecology of anthrax. Science 172:1303–1307, 1971.Wade, N. Death at Sverdlovsk: A critical diagnosis. Science 209:1501–1502, 1980.Whitford, H.W. Anthrax. In: Steele, J.H., H. S<strong>to</strong>enner, W. Kaplan, M. Torten, eds. Vol I,Section A: CRC H<strong>and</strong>book Series in Zoonoses. Boca Ra<strong>to</strong>n: CRC Press; 1979.Wright, G.G. Anthrax. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger, eds.Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.


BOTULISMICD-10 A05.1Synonyms: Allantiasis; “lamziekte” (bovine botulism in South Africa); “limberneck”(botulism in fowl).Etiology: Toxins produced by Clostridium botulinum, which are the most potentknown. C. botulinum is an obligate, spore-forming anaerobe. It has been sub-classified(Smith, 1977) in<strong>to</strong> four groups (I <strong>to</strong> IV), according <strong>to</strong> culture <strong>and</strong> serologicalcharacteristics. Seven different types of botulinum antigens have been identified(A–G), according <strong>to</strong> their serological specificity. Classical botulism results from


BOTULISM 29preformed <strong>to</strong>xins ingested with food. In wound botulism, the <strong>to</strong>xin forms by contaminationof the injured tissue. In 1976, a new clinical type, infant botulism, wasidentified. It is caused by colonization of the infant’s intestinal tract by C. botulinum<strong>and</strong> the resultant production <strong>and</strong> absorption of <strong>to</strong>xins.The species C. botulinum is very heterogeneous. The different groups (I <strong>to</strong> IV) aredifferentiated according <strong>to</strong> their ability <strong>to</strong> digest proteins <strong>and</strong> break down sugars.Group I is highly proteolytic <strong>and</strong> saccharolytic <strong>and</strong> includes all the type A strains aswell as various type B <strong>and</strong> F strains. Group II includes all the type E strains <strong>and</strong> thenonproteolytic type B <strong>and</strong> F strains that are highly saccharolytic. Group III consistsof the type C <strong>and</strong> D strains, which are not proteolytic (except that they digest gelatin).Group IV contains only type G, which is proteolytic but not saccharolytic(Sakaguchi et al., 1981; Concon, 1988).Despite the metabolic <strong>and</strong> DNA differences among them, these groups ofclostridia have until now been classified in a single species because they all producea botulinum neuro<strong>to</strong>xin that acts similarly in animal hosts (Ca<strong>to</strong> et al., 1986).However, not all researchers agree with this scheme <strong>and</strong> one argument of thosefavoring a reclassification is the recent discovery of neuro<strong>to</strong>xigenic strains inClostridium baratii <strong>and</strong> C. butyricum.In effect, two cases of type E infant botulism in Rome, Italy, caused by C.butyricum (Aureli et al., 1986) have been described <strong>and</strong> another case was describedin New Mexico (USA) in a child suffering from a neuro<strong>to</strong>xigenic type F botulismproduced by a clostridium that was later identified as C. baratii (Hall et al., 1985).These clostridia were identified on the basis of their phenotype characteristics <strong>and</strong>were later confirmed through DNA hybridization (Suen et al., 1988a). The neuro<strong>to</strong>xinisolated from C. baratii was similar in structure <strong>and</strong> amino acid sequence <strong>to</strong>C. botulinum types A, B, <strong>and</strong> E (Giménez et al., 1992). The proponents of reclassification,such as Suen et al. (1988b), have suggested renaming group IV, which containsthe single <strong>to</strong>xigenic type G, as Clostridium argentinense. Arnon (1986) agreesthat reclassification would be justified based on logical criteria <strong>and</strong> taxonomicpurity, but questions whether this would improve clinical, microbiological, <strong>and</strong> epidemiologicalknowledge.Geographic Distribution: Occurs on all continents, with a marked regional distributionthat probably reflects the presence in the soil of the microorganism <strong>and</strong> itsdifferent types of <strong>to</strong>xins.Occurrence in Man: The disease occurs more frequently in the northern hemispherethan in the southern hemisphere, <strong>and</strong> can appear sporadically <strong>and</strong> amonggroups of people who ingest the same food with the preformed <strong>to</strong>xin. From 1950 <strong>to</strong>1973, an average of 15.1 outbreaks occurred annually in the US, with 2.4 cases peroutbreak. In that period, there were only three outbreaks affecting more than 10 people,but in 1977, an outbreak of 59 cases involving type B botulinum <strong>to</strong>xin wasdescribed, caused by food eaten in a restaurant (Terranova et al., 1978). Figure 3illustrates the reported cases <strong>and</strong> deaths by year in the US during the period1960–1980 (<strong>PAHO</strong>, 1982). More than half of the cases reported since 1899 from 45states occurred in five western states. Table 1 shows the foods <strong>and</strong> type of botulinum<strong>to</strong>xin that caused the illness. In the United States, only 4% of the outbreaks originatedin restaurants, but they represent 42% of the 308 cases occurring between1976 <strong>and</strong> 1984. The most widespread outbreak recorded in the United States


30 BACTERIOSESFigure 3. Botulism (transmitted by foods). Reported cases <strong>and</strong> deaths per year, United States of America, 1960–1980.10090807060Number50403020101960 '62 '64 '66 '68 '70 '72 '74 '76 '78 '80YearCasesDeaths (data not available for 1979 <strong>and</strong> 1980)Source: <strong>PAHO</strong> Epidemiol Bull 3(4):2, 1982.occurred in Michigan in 1977, when 59 people became sick after eating restaurantfood that had been home-canned <strong>and</strong> contained botulinum <strong>to</strong>xin B (MacDonald etal., 1986). In Canada, between 1979 <strong>and</strong> 1980, 15 incidents were investigated(<strong>PAHO</strong>, 1982). In Argentina, during the period 1967–1981, 139 cases were reported(Figure 4). In 1958, several suspected cases of botulism occurred in Brazil when sixmembers of the same family died <strong>and</strong> others became ill after eating home-cannedboiled fish. In 1981, two other suspected cases occurred in Rio de Janeiro, causedby ingestion of an industrially processed food.In Europe <strong>and</strong> Asia, the occurrence of the illness varies from one country <strong>to</strong>another. In Pol<strong>and</strong>, which seems <strong>to</strong> be the country hardest hit by botulism, themajority of cases have occurred in rural areas, as they have in other countries. From1979 <strong>to</strong> 1983, there were a <strong>to</strong>tal of 2,390 cases <strong>and</strong> 45 deaths, with an average of478 cases per year (Hauschild, 1989). The largest outbreak known <strong>to</strong> date <strong>to</strong>ok placein Dniepropetrovsk (former USSR), in 1933, with 230 cases <strong>and</strong> 94 deaths. Morerecently, about 14 outbreaks per year have occurred. In France, botulism is infrequent,with about four outbreaks annually. However, during World War II, 500 outbreakswere recorded with more than 1,000 cases in that country. Germany is second<strong>to</strong> Pol<strong>and</strong> in the incidence of the illness. In the rest of Europe botulism is rare.During the period 1958–1983, there were 986 outbreaks, 4,377 cases, <strong>and</strong> 548deaths in China; most of the cases (81%) occurred in the northwest province ofXinjiang (Ying et al., 1986). During the period 1951–1984, there were 96 outbreaks,


Table 1. Foods giving rise <strong>to</strong> botulism, <strong>and</strong> number of outbreaks, United States of America, 1899–1977. a, bType of Fish <strong>and</strong> Milk <strong>and</strong>botulinum fish Condi- milk<strong>to</strong>xin Vegetables products Fruits ments c Beef d products Pork Fowl Others e Unknown e TotalA 115 11 22 17 6 3 2 2 8 9 195B 31 4 7 5 1 2 1 2 3 3 59E 1 25 — — — — — — 3 1 30F — — — — 1 — — — — — 1A <strong>and</strong> B 2 — — — — — — — — — 2Unknown e 2 1 — 1 — — — — — 6 10Total 151 41 29 23 8 5 3 4 14 19 297a For the period 1899–1973, only outbreaks in which the type of <strong>to</strong>xin was confirmed are included; for 1974–1977, all outbreaks are included.b Prepared by the Centers for Disease Control <strong>and</strong> Prevention, Atlanta, Georgia, USA.c Includes outbreaks caused by <strong>to</strong>ma<strong>to</strong> condiments, hot sauces, <strong>and</strong> salad dressings.d Includes one type F outbreak caused by venison, <strong>and</strong> one type A outbreak caused by mut<strong>to</strong>n.e Categories added for the period 1974–1977.Source: <strong>PAHO</strong> Epidemiol Bull 3(4):2, 1982.


32 BACTERIOSESFigure 4. Reported cases of botulism per year, Argentina, 1967–1981.30272421Number of cases18151296301967 '68 '69 '70 '71 '72 '73 '74 '75 '76 '77YearSource: <strong>PAHO</strong> Epidemiol Bull 3(4):2, 1982.'78 '79 '80 '81478 cases, <strong>and</strong> 109 deaths in Japan (Hauschild, 1989). In the rest of Asia <strong>and</strong> inAfrica, few cases have been identified (Smith, 1977).Infant botulism was recognized for the first time in the United States, <strong>and</strong> then inCanada, several European countries, <strong>and</strong> Australia. From 1976 <strong>to</strong> 1980, 184 caseswere recorded in the United States, 88 of which occurred in California <strong>and</strong> 96 in 25other states. Currently, infant botulism is the predominant form in the United States,with approximately 100 cases reported each year (Suen et al., 1988). The diseasehas also been described in Argentina, Australia, Canada, the former Czechoslovakia,Great Britain, <strong>and</strong> Italy. Almost all cases occurred in children under 6 months of age(Morris, 1983) though some occurred in children up <strong>to</strong> 1 year.The first case of wound botulism was recognized in the United States in 1943. By1982, 27 cases were recorded, 20 of them in states west of the Mississippi River(CDC, 1982). In 1986, MacDonald et al. reported on the incidence of botulism inthe United States between 1976 <strong>and</strong> 1984, including 16 cases of wound botulism(two of these in drug addicts using intravenous drugs).Occurrence in Animals: Botulism in animals, including birds, is caused by typesC (C alpha <strong>and</strong> C beta) <strong>and</strong> D, but there are also outbreaks due <strong>to</strong> A, E, <strong>and</strong> B.Botulism in bovines is becoming economically important in some areas, where itcan affect a large number of animals. Such areas, generally poor in phosphorus, arefound in the southwestern United States, in Corrientes province in Argentina, <strong>and</strong> inPiaui <strong>and</strong> Mat<strong>to</strong> Grosso in Brazil. However, bovine botulism occurs even more frequentlyin South Africa (“lamziekte”) <strong>and</strong> Australia. It is also important in Senegal,where it is believed <strong>to</strong> cause more cattle loss than any other disease. In other countries,sporadic outbreaks <strong>and</strong> cases occur, primarily caused by ingestion of fodder


BOTULISM 33<strong>and</strong> silage containing the preformed <strong>to</strong>xins (Smith, 1977). Recently, various importan<strong>to</strong>utbreaks have been recorded due <strong>to</strong> consumption of bedding silage <strong>and</strong> birddroppings. In Great Britain, 80 out of 150 stabled cattle became sick <strong>and</strong> 68 died.Type C <strong>to</strong>xin was detected in 18 of the 22 sera examined <strong>and</strong> the same <strong>to</strong>xin wasconfirmed in the remains of dead chickens found in the silage (McLoughlin et al.,1988). Similar outbreaks also occurred in Brazil <strong>and</strong> Canada because the bird beddingused <strong>to</strong> feed the cattle contained the type C <strong>to</strong>xin (Bienvenue et al., 1990;Schocken-Iturrino et al., 1990). Animal remains are usually found when botulismoutbreaks occur in cattle, especially due <strong>to</strong> silage; however, no animal remains couldbe found in the fodder in various cases in the United States <strong>and</strong> Europe.Bovine poisoning by type B is rare; outbreaks have occurred in Europe (Blood etal., 1983), the United States (Divers et al., 1986), <strong>and</strong> Brazil (Loba<strong>to</strong> et al., 1988).Botulism in sheep is due <strong>to</strong> type C <strong>and</strong> has been identified only in westernAustralia <strong>and</strong> South Africa.In horses, botulism cases are sporadic <strong>and</strong> for the most part are caused by C.botulinum type C. It has been diagnosed in various European countries, the US,Israel, Australia, <strong>and</strong> South Africa. A special form occurs in colts at 6 <strong>to</strong> 8 weeksold (“shaker foal syndrome”); it is due <strong>to</strong> the type B neuro<strong>to</strong>xin <strong>and</strong> its pathogenesisis similar <strong>to</strong> botulism in children, since apparently C. botulinum has <strong>to</strong> colonizefirst in the intestine <strong>and</strong> other sites in order <strong>to</strong> produce the neuro<strong>to</strong>xin later.Outbreaks of this form have been described in the United States <strong>and</strong> Australia(Thomas et al., 1988).Botulism in swine is rare because of the natural high resistance of this species <strong>to</strong>botulinum <strong>to</strong>xin. Outbreaks diagnosed in Senegal <strong>and</strong> Australia were caused by typeC beta <strong>and</strong> one in the United States was caused by type B.Botulism in mink can be an important problem owing <strong>to</strong> their eating habits, if theyare not vaccinated as recommended. Mink are highly susceptible <strong>to</strong> type C, whichcauses almost all the outbreaks.Botulism in fowl occurs practically worldwide <strong>and</strong> is caused principally by typeC alpha. Outbreaks of types A <strong>and</strong> E have been recorded in waterfowl. In the westernUnited States, type C is responsible for massive outbreaks in wild ducks duringthe summer <strong>and</strong> early fall. Many other species of wild fowl are susceptible <strong>to</strong> botulism<strong>and</strong> outbreaks also occur in domestic chickens <strong>and</strong> farm-bred pheasants (Smith,1977). Botulism in domestic fowl has been related <strong>to</strong> cannibalism <strong>and</strong> the ingestionof maggots in decomposing carcasses. The explanation is that the body temperatureof fowl (41°C) favors the type C <strong>to</strong>xin, which would be produced <strong>and</strong> absorbed inthe caecum, whose pH of 7.4 also favors the <strong>to</strong>xin (Castro et al., 1988).Botulism in dogs is rare <strong>and</strong> is caused primarily by ingesting bird carcasses, withtype C being responsible for the disease (Hatheway <strong>and</strong> McCroskey, 1981).The Disease in Man: (a) Botulism poisoning by foods is produced primarily bytypes A, B, <strong>and</strong> E <strong>and</strong> rarely by F or G. Outbreaks in man described as type C havenot been confirmed, as the <strong>to</strong>xin has not been found in the patients’ blood or fecesnor in foods they ate. An outbreak of type D was identified in Chad, Africa, amongpeople who had eaten raw ham (Smith, 1977). A new <strong>to</strong>xicogenic type, C. botulinumtype G, was isolated from the soil in Argentina in 1969 (Giménez <strong>and</strong> Ciccarelli,1970). The first human cases were recognized in Switzerl<strong>and</strong> (Sonnabend et al.,1981). The microorganism was isolated in au<strong>to</strong>psy specimens from four adults <strong>and</strong>


34 BACTERIOSESan 18-week-old child. In addition, the presence of the <strong>to</strong>xin could be confirmed inthe blood serum of three of these persons who died suddenly at home. The symp<strong>to</strong>mswere similar <strong>to</strong> those of classic botulism. Nine additional cases of sudden <strong>and</strong>unexpected death were described (Sonnabend et al., 1985).The incubation period is usually from 18 <strong>to</strong> 36 hours, but the illness can show upwithin a few hours or as long as eight days after ingestion of the contaminated food.The clinical signs of the different types vary little, although the mortality rate seems<strong>to</strong> be higher for type A. The disease is afebrile, <strong>and</strong> gastrointestinal symp<strong>to</strong>ms, suchas nausea, vomiting, <strong>and</strong> abdominal pain, precede neurological symp<strong>to</strong>ms.Neurological manifestations are always symmetrical, with weakness or descendingparalysis. Diplopia, dysarthria, <strong>and</strong> dysphagia are <strong>common</strong>. Consciousness <strong>and</strong> sensibilityremain intact until death. The immediate cause of death is usually respira<strong>to</strong>ryfailure. The mortality rate in botulinum poisonings is high. The highest rate isrecorded in patients with short incubation periods, i.e., those who have ingested ahigh dose of the <strong>to</strong>xin. In the United States, the fatality rate has been reduced from60% in 1899–1949 <strong>to</strong> 15.7% in 1970–1977, by means of early <strong>and</strong> proper treatment.In patients who survive, complete recovery, especially of ocular movement, maytake as long as six <strong>to</strong> eight months.Treatment should be initiated as soon as possible through administration of thetrivalent botulinum anti<strong>to</strong>xin (A, B, <strong>and</strong> E). The patient must be hospitalized inintensive care in order <strong>to</strong> anticipate <strong>and</strong> treat respira<strong>to</strong>ry distress, which is the immediatecause of death (Benenson, 1990).(b) Infant botulism is an intestinal infection caused by the ingestion of C. botulinumspores, which change in the intestine in<strong>to</strong> the vegetative form, multiply, <strong>and</strong>produce <strong>to</strong>xins. Of 96 cases studied (Morris et al., 1983) in the United States,excluding California, 41 were caused by C. botulinum type A, 53 by type B, one bytype F, <strong>and</strong> another by type B <strong>to</strong>gether with F. Type A appeared almost exclusivelyin the western states, while type B predominated in the East. This distribution is similar<strong>to</strong> that of the spores in the environment (see the section on source of poisoningor infection <strong>and</strong> mode of transmission). In addition, two cases in Italy due <strong>to</strong> type Eproduced by Clostridium butyricum <strong>and</strong> one case in New Mexico (USA) due <strong>to</strong> typeF produced by Clostridium baratii have been described; i.e., by two species otherthan C. botulinum (see section on etiology). The case of a girl under age 6 monthswith paroxystic dyspnea due <strong>to</strong> C. botulinum type C <strong>to</strong>xin was also described. Thechild survived the illness, but may have been left with a cerebral lesion probablycaused by hypoxia (Oguma et al., 1990). Sonnabend et al. (1985) had previouslyidentified the bacteria <strong>and</strong> C <strong>to</strong>xin in the colon of a child who died suddenly inSwitzerl<strong>and</strong>.The fact that C. botulinum primarily colonizes the caecum <strong>and</strong> colon, <strong>and</strong> that96% of patients with infant botulism are under 6 months of age led <strong>to</strong> research onthe characteristics of the intestinal microflora that allow the clostridium <strong>to</strong> multiply.Using a normal mouse as a model, it was established that the microflora in adultsprevent the establishment of C. botulinum; however, if adult “germ-free” mice areused, the clostridium multiplies in their intestines. The same thing happens <strong>to</strong> conventionalmice from 7 <strong>to</strong> 13 days old, which are very susceptible. In addition, breastfedinfants have feces with a lower pH (5.1–5.4) than those fed with formula (pH5.9–8.0). This difference may be significant, because the multiplication of C. botulinumdeclines as pH falls <strong>and</strong> ceases below 4.6. In addition, breast-feeding has the


BOTULISM 35advantage of transferring immunogenic fac<strong>to</strong>rs <strong>to</strong> the infant (Arnon, 1986). Anotherimportant fac<strong>to</strong>r seems <strong>to</strong> be the frequency of defecation: of 58 patients who wereat least 1 month old, 37 (64%) had a usual pattern of one defecation per day, as compared<strong>to</strong> 17 (15%) of the 115 in the control group (Spika et al., 1989).In adults, botulism may occur without the presence of preformed <strong>to</strong>xin in food: itmay occur through colonization of the large intestine by C. botulinum, the productionof <strong>to</strong>xins, <strong>and</strong> their absorption. Such cases are rare <strong>and</strong> primarily affect thosewho have alterations in intestinal structure <strong>and</strong> microflora.The disease in infants begins with constipation, followed by lethargy <strong>and</strong> loss ofappetite, p<strong>to</strong>sis, difficulty swallowing, muscular weakness, <strong>and</strong> loss of head control.Neuromuscular paralysis may progress from the cranial nerves <strong>to</strong> the peripheral <strong>and</strong>respira<strong>to</strong>ry muscles, resulting in death. The severity of the illness varies from moderate<strong>to</strong> life-threatening, causing sudden death of the child. It has been estimated thatinfant botulism is responsible for at least 5% of the cases of sudden infant death syndrome(Benenson, 1990).(c) Wound botulism is clinically similar <strong>to</strong> classic botulism in its neurologicalsyndrome. It is a <strong>to</strong>xin infection produced as the result of a contaminated wound thatcreates anaerobic conditions where C. botulinum can become established, reproduce,<strong>and</strong> develop a neuro<strong>to</strong>xin that is absorbed by the vessels. Of the 27 knowncases, 15 were associated with type A, 5 with type B, 1 with both A <strong>and</strong> B, <strong>and</strong> onewas undetermined.The Disease in Animals: Botulism in domestic mammals is caused primarily bytypes C <strong>and</strong> D, <strong>and</strong> in fowl, by type C. Outbreaks in bovines are usually associatedwith a phosphorus deficiency <strong>and</strong> the resultant osteophagea <strong>and</strong> compulsive consumption(“pica”) of carrion containing botulinum <strong>to</strong>xins. In locations where typesC beta <strong>and</strong> D are found, such as South Africa, C. botulinum spores multiply rapidlyin carrion <strong>and</strong> produce <strong>to</strong>xins <strong>to</strong> which bovines are very susceptible. The mainsymp<strong>to</strong>m is the partial or complete paralysis of the locomo<strong>to</strong>r, mastica<strong>to</strong>ry, <strong>and</strong>swallowing muscles. The animals have difficulty moving, stay motionless or recumbentfor long periods of time, <strong>and</strong>, as the illness progresses, cannot hold their headsup <strong>and</strong> so bend their necks over their flanks. The mortality rate is high.In sheep, botulism is associated with protein <strong>and</strong> carbohydrate deficiencies, whichlead the animals <strong>to</strong> eat the carcasses of small animals they find as they graze.In horses, as in other mammals, the incubation period varies widely according <strong>to</strong>the amount of <strong>to</strong>xin ingested. In very acute cases, death may ensue in one or twodays. When the course is slower, the disease generally begins with paralysis of thehind quarters <strong>and</strong> progresses <strong>to</strong> other regions of the body until it produces death due<strong>to</strong> respira<strong>to</strong>ry failure. A <strong>to</strong>xin infection similar <strong>to</strong> infant botulism <strong>and</strong> wound botulismhas been described in young colts. The neuromuscular paralytic syndromeaffects colts from 2 <strong>to</strong> 8 weeks old; they show signs of progressive mo<strong>to</strong>r paralysisthat includes muscular tremors (shaker foal syndrome), dysphagia with flaccidparalysis of the <strong>to</strong>ngue, difficulty remaining on their feet, a tendency <strong>to</strong> collapse,dyspnea, mydriasis, <strong>and</strong> constipation (Thomas et al., 1988). Sometimes they diewithout signs of disease (“sudden death”). The disease is produced by the type B<strong>to</strong>xin that requires prior colonization of C. botulinum in gastric, intestinal, umbilical,hepatic (necrosis), muscular, or subcutaneous lesions. Necrotic lesions seem <strong>to</strong>be necessary for <strong>to</strong>xicity, as in wound botulism in man (Swerczek, 1980).


36 BACTERIOSESOutbreaks with high death rates have been observed on mink farms. Food poisoningin these animals is due primarily <strong>to</strong> type C beta.In ducks <strong>and</strong> other waterfowl, the first symp<strong>to</strong>m of poisoning is paralysis of thewings, which then extends <strong>to</strong> other muscles, <strong>and</strong> finally <strong>to</strong> those of the neck. Thebirds drown when they can no longer hold their heads above water. An outbreak due<strong>to</strong> type E occurred in waterfowl on Lake Michigan (USA).The illness in chickens is produced mainly by type C alpha. It has been giventhe name “limberneck” because flaccid paralysis is frequently observed in afflictedbirds.Treatment with botulinum anti<strong>to</strong>xin produced variable results in bovines. Betterresults were obtained from anti<strong>to</strong>xin C in mink <strong>and</strong> ducks, but the cost can be excessive.Control <strong>and</strong> prevention must be the principal <strong>to</strong>ols for combating losses due <strong>to</strong>animal botulism.Source of Poisoning or Infection <strong>and</strong> Mode of Transmission: The reservoir ofC. botulinum is the soil, river <strong>and</strong> sea sediments, vegetables, <strong>and</strong> the intestinal tractsof mammals <strong>and</strong> birds. The spores formed by the bacteria are very resistant <strong>to</strong> heat<strong>and</strong> desiccation. The etiologic agent is distributed on all continents, though irregularly.The distribution of the <strong>to</strong>xicogenic types also varies according <strong>to</strong> region. In astudy (Smith, 1978) carried out across the United States, subdividing it in<strong>to</strong> fourtransverse sections, C. botulinum was found in 23.5% of 260 soil samples. Type Awas most prevalent in the western states with neutral or alkaline soil. Type B had amore uniform distribution, but predominated in the East, a pattern which seems <strong>to</strong>be associated with highly organic soils. Type C was found in acid soils on the GulfCoast, type D in some alkaline soils in the West, <strong>and</strong> type E in the humid soils ofseveral states. In the former Soviet Union, C. botulinum was isolated from 10.5% of4,242 soil samples, with type E accounting for 61% of all positive cultures. Thegreatest concentration of spores was found in the European section of the countrysouth of 52° N latitude (Kravchenko <strong>and</strong> Shishulina, 1967).The wide distribution of C. botulinum in nature explains its presence in food.Vegetables are contaminated directly from the soil. Foods of animal origin are probablycontaminated via the animals’ intestinal tract <strong>and</strong> by spores in the environment.The main source of botulinum poisoning for man <strong>and</strong> animals is food in which themicroorganism has multiplied <strong>and</strong> produced the powerful <strong>to</strong>xin. After ingestion, the<strong>to</strong>xin is absorbed through the intestine, primarily the upper portion, <strong>and</strong> carried bythe bloodstream <strong>to</strong> the nerves. It acts upon the presynaptic union of cholinergicnerve endings by inhibiting the release of acetylcholine.Any food, whether of vegetable or animal origin, can give rise <strong>to</strong> botulism if conditionsfavor the multiplication of C. botulinum <strong>and</strong>, consequently, the production of<strong>to</strong>xins. The main requirements for the multiplication of C. botulinum are anaerobiosis<strong>and</strong> a pH above 4.5, but once the <strong>to</strong>xin is formed an acid medium can be favorable.Home-canned foods are generally responsible for the disease, although incorrectlysterilized or preserved commercial products are sometimes the cause.Poisoning ensues after eating a raw or insufficiently cooked product that was preservedsome time earlier. The types of food responsible for poisoning vary according<strong>to</strong> regional eating habits.The most <strong>common</strong> sources of types A <strong>and</strong> B botulism in the United States <strong>and</strong>Canada are home-canned fruits <strong>and</strong> vegetables. In Europe, on the other h<strong>and</strong>, meat


BOTULISM 37<strong>and</strong> meat products seem <strong>to</strong> play the most important role. In Japan, the former SovietUnion, the northern United States, Alaska, <strong>and</strong> northern Canada, type E, which isassociated with foods of marine origin, predominates.Ethnic cus<strong>to</strong>ms <strong>and</strong> food habits often favor food poisoning. In 1992, a family ofEgyptian origin living in New Jersey (USA) became sick from type E botulismafter consuming “moloha,” an uneviscerated salt-cured fish (CDC, 1992). There hadbeen an earlier outbreak in which two Russian immigrants in New York <strong>and</strong> fivepeople in Israel became ill with type E botulism; all of them had eaten unevisceratedsalt-cured, air-dried fish (“ribyetz”) from the same source (CDC, 1989). A preservedregional dish that is popular in the Orient, a soft cheese prepared with soymilk, led <strong>to</strong> 705 (71.5%) of the 986 outbreaks of botulism recorded in China (Ying<strong>and</strong> Shuan, 1986).In contrast <strong>to</strong> classic botulism (acquired through foods), infant botulism begins asan intestinal infection caused by C. botulinum, where the spores germinate, multiply,<strong>and</strong> produce the <strong>to</strong>xin that is absorbed through the intestinal wall. Honey hasbeen implicated as a source of infection, since it is frequently a supplementary foodfor the nursing child. However, results of research on the presence of botulinumspores in this food, as well as epidemiological investigations, are inconclusive. Inany case, there is no doubt that the infection is caused by ingestion.The source of wound botulism is environmental. Botulism in bovines results fromgrazing (“lamziekte”) or the consumption of bailed fodder or silage. Poisoning contractedwhile grazing usually occurs in areas lacking or deficient in phosphorus.Many species of animals in the area contain C. botulinum in their intestinal flora;when an animal dies, these bacteria invade the whole organism <strong>and</strong> produce greatquantities of <strong>to</strong>xin. Bovines suffering from pica ingest animal remains containingthe preformed <strong>to</strong>xin <strong>and</strong> contract botulism. After dying, these same bovines are asource of poisoning for the rest of the herd. Mortality in cattle has also been ascribed<strong>to</strong> drinking water that contained the decomposed bodies of small animals. Botulismcontracted through the consumption of fodder or silage is produced by the accidentalpresence of a small animal’s body (usually a cat) or the remains of birds <strong>and</strong> thediffusion of the botulinum <strong>to</strong>xin around them in<strong>to</strong> the food (Smith, 1977). Thesources of poisoning for other mammalian species are similar.For wild ducks, the source of poisoning is insect larvae that invade the bodies ofducks that died from various causes. If a duck had C. botulinum in its intestinal flora,the bacteria invades the whole organism after its death. The larvae then absorb the<strong>to</strong>xin produced, constituting a source of <strong>to</strong>xin for birds. It is estimated that a duckneed only ingest a few such larvae for death by botulism <strong>to</strong> ensue.Research on outbreaks among pheasants has found that they ate maggots from thebodies of small animals.Role of Animals in the Epidemiology of the Disease: No epidemiological relationshipbetween human <strong>and</strong> animal botulism has been established. C. botulinumtype A spores have been isolated from animal feces, <strong>and</strong> types A <strong>and</strong> B botulismmicroorganisms have been found in the intestine <strong>and</strong> liver of bovines that died fromother causes. The microorganism has also been isolated from the intestine <strong>and</strong> bonemarrow of healthy dogs. Thus the possibility exists that these animals are carriers ofthe microorganism <strong>and</strong> serve <strong>to</strong> transport <strong>and</strong> disseminate C. botulinum from oneplace <strong>to</strong> another.


38 BACTERIOSESDiagnosis: Clinical diagnosis should be confirmed with labora<strong>to</strong>ry tests. Themost conclusive evidence is the presence of botulinum <strong>to</strong>xin in the serum of thepatient. S<strong>to</strong>mach contents <strong>and</strong> fecal material of persons exposed <strong>to</strong> the suspectedfood should also be examined for the <strong>to</strong>xin. The food in question should be cultured<strong>to</strong> isolate <strong>and</strong> identify the microorganism. In infant botulism, the attempt is made <strong>to</strong>isolate the agent <strong>and</strong> the <strong>to</strong>xin from the infant’s feces, since the <strong>to</strong>xin is rarelydetected in serum. An enzyme-linked immunosorbent assay (ELISA) test has beendeveloped for the detection of A <strong>and</strong> B <strong>to</strong>xin in children’s fecal samples; this maybe useful as a screening test in clinical specimens (Dezfulian et al., 1984). When awound is the suspected origin of the poisoning, fluid is aspirated from the wound<strong>and</strong> biopsies are performed for bacteriological examination.Control: With regard <strong>to</strong> man, control measures include: (a) regulation <strong>and</strong> inspectionof industrial bottling, canning, <strong>and</strong> food-preserving processes, <strong>and</strong> (b) healtheducation <strong>to</strong> point out the dangers of home canning <strong>and</strong> <strong>to</strong> make the public aware ofimportant fac<strong>to</strong>rs in the preservation of home products, such as duration, pressure,<strong>and</strong> temperature of sterilization. Home-canned foods should be boiled before beingserved <strong>to</strong> destroy the <strong>to</strong>xins that are thermolabile. Foods from swollen cans or foodaltered in taste, smell, or appearance should not be eaten even after cooking. Anyfood that is bottled, canned, or prepared in some other way (salted, dried, etc.) <strong>and</strong>has led <strong>to</strong> a case or outbreak should be seized.Immediate epidemiological investigation <strong>and</strong> prompt diagnosis of an outbreak areessential <strong>to</strong> both the prevention of new cases <strong>and</strong> the recovery of the patient.In areas where botulism in animals is a problem, the diet of lives<strong>to</strong>ck should besupplemented with feed rich in phosphorus <strong>to</strong> avoid osteophagia or pica; vaccinationof animals with the appropriate <strong>to</strong>xoid can yield good results. When bird beddingis used as silage for cattle or for pasture fertilizer, any remains of birds or otheranimals should be carefully eliminated. When an outbreak occurs in a fowl facility,carcasses must be removed as soon as possible <strong>to</strong> prevent its progression.BibliographyArnon, S.S. Infant botulism: Anticipating the second decade. J Infect Dis 154:201–206, 1986.Aureli, P.L., F.B. Pasolini, M. Gianfranceschi, et al. Two cases of type E infant botulismcaused by neuro<strong>to</strong>xigenic Clostridium butyricum in Italy. J Infect Dis 154:207–211, 1986.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bienvenue, J.G., M. Morin, S. Forget. Poultry litter associated botulism (type C) in cattle.Can Vet J 31:711, 1990.Blood, D.C., O.M. Radostits, J.A. Henderson. Veterinary Medicine. 6th ed. London:Baillière Tindall; 1983.Castro, A.G.M., A.M. Carvalho, L. Baldassi, et al. Botulismo em aves de postura no estadode São Paulo. Arq Inst Biol 55:1–4, 1988.Ca<strong>to</strong>, E.P., W.L. George, S.N. Finegold. Clostridium. In: Sneath, P.H.A., N.S. Mair, M.E.Sharpe, J.G. Holt, eds. Bergey’s Manual of Systemic Bacteriology. Vol 2. Baltimore: Williams& Wilkins; 1986.Ciccarelli, A.S., D.F. Giménez. Clinical <strong>and</strong> epidemiological aspects of botulism inArgentina. In: Lewis, G.E., Jr., ed. Biomedical Aspects of Botulism. New York: AcademicPress; 1981.


BOTULISM 39Concon, J.M. Part B: Contaminants <strong>and</strong> additives. In: Food Toxicology. New York: MarcelDekker; 1988.Dezfulian, M., C.L. Hatheway, R.H. Yolken, J.G. Bartlett. Enzyme-linked immunosorbentassay for detection of Clostridium botulinum type A <strong>and</strong> type B <strong>to</strong>xins in s<strong>to</strong>ol samples ofinfants with botulism. J Clin Microbiol 20:379–383, 1984.Divers, T.J., R.C. Bartholomew, J.B. Messick, et al. Clostridium botulinum type B <strong>to</strong>xicosisin a herd of cattle <strong>and</strong> a group of mules. J Am Vet Med Assoc 188:382–386, 1986.Giménez, D.F., A.S. Ciccarelli. Another type of Clostridium botulinum. Zentralbl Bakteriol215:221–224, 1970.Giménez, D.F., A.S. Ciccarelli. Clostridium botulinum en Argentina: presente y futuro. RevAsoc Argent Microbiol 8:82–91, 1976.Giménez, J.A., M.A. Giménez, B.R. Dasgupta. Characterization of the neuro<strong>to</strong>xin from aClostridium baratii strain implicated in infant botulism. Infect Immun 60:518–522, 1992.Hall, J.D., L.M. McCroskey, B.J. Pincomb, C.L. Hatheway. Isolation of an organism resemblingClostridium barati which produces type F botulinal <strong>to</strong>xin from an infant with botulism.J Clin Microbiol 21(4):654–655, 1985.Hatheway, C.L., L.M. McCroskey. Labora<strong>to</strong>ry investigation of human <strong>and</strong> animal botulism.In: Lewis, G.E., Jr., ed. Biomedical Aspects of Botulism. New York: Academic Press; 1981.Hauschild, A.H. Clostridium botulinum. In: Doyle, M.P., ed. Foodborne BacterialPathogens. New York: Marcel Dekker; 1989.Ingram, M., T.A. Roberts. Botulism 1966. London: Chapman & Hall; 1967.Kravchenko, A.T., L.M. Shishulina. Distribution of C. botulinum in soil <strong>and</strong> water in theUSSR. In: Ingram, M., T.A. Roberts, eds. Botulism 1966. London: Chapman & Hall; 1967.Loba<strong>to</strong>, F.C.F., M.A. de Melo, N. de Silva, J.M. Diniz. Botulismo em bovinos causado peloClostridium botulinum tipo B [letter]. Arq Brasil Med Vet Zootec 40:445–446, 1988.MacDonald, K.L., M.L. Cohen, P.A. Blake. The changing epidemiology of adult botulismin the United States. Am J Epidemiol 124:794–799, 1986.McLoughlin, M.F., S.G. McIlroy, S.D. Neill. A major outbreak of botulism in cattle beingfed ensiled poultry litter. Vet Rec 122:579–581, 1988.Morris, J.G., J.D. Snyder, R. Wilson, R.A. Feldman. Infant botulism in the United States:An epidemiological study of cases occurring outside of California. Am J Public Health73:1385–1388, 1983.Oguma, K., K. Yokota, S. Hyashi, et al. Infant botulism due <strong>to</strong> Clostridium botulinum typeC <strong>to</strong>xin. Lancet 336:1449–1450, 1990.Pan American Health Organization (<strong>PAHO</strong>). Botulism in the Americas. Epidemiol Bull3(4):1–3, 1982.Prévot, A.R., A. Turpin, P. Kaiser. Les bactéries anaérobies. Paris: Dunod; 1967.Riemann, H. Botulism: Types A, B, <strong>and</strong> F. In: Riemann, H., ed. Food-borne Infections <strong>and</strong>In<strong>to</strong>xications. New York: Academic Press; 1969.Sakaguchi, G., I. Ohishi, S. Kozaki. Purification <strong>and</strong> oral <strong>to</strong>xicities of Clostridium botulinumprogeni<strong>to</strong>r <strong>to</strong>xins. In: Lewis, G.E., Jr., ed. Biomedical Aspects of Botulism. New York:Academic Press; 1981.Schocken-Iturrino, R.P., F.A. Avila, S.C.P. Berchielli, et al. Cama de frango contaminadacon <strong>to</strong>xina botulínica. Ciencia Vet Jaboticabal 4:11–12, 1990.Smith, L.D. Clostridial <strong>diseases</strong> of animals. Adv Vet Sci 3:463–524, 1957.Smith, L.D. Botulism: The Organism, its Toxins, the Disease. Springfield: Thomas; 1977.Smith, L.D. The occurrence of Clostridium botulinum <strong>and</strong> Clostridium tetani in the soil ofthe United States. Health Lab Sci 15:74–80, 1978.Sonnabend, O., W. Sonnabend, R. Heinzle, T. Sigrist, R. Dirnohofer, U. Krech. Isolation ofClostridium botulinum type G <strong>and</strong> identification of type G botulinal <strong>to</strong>xin in humans: Repor<strong>to</strong>f five sudden unexpected deaths. J Infect Dis 143:22–27, 1981.Sonnabend, O.A., W.F. Sonnabend, U. Krech, et al. Continuous microbiological <strong>and</strong> patho-


40 BACTERIOSESlogical study on 70 sudden <strong>and</strong> unexpected infant deaths: Toxigenic intestinal Clostridiumbotulinum infection in 9 cases of sudden infant death syndrome. Lancet 2:237–241, 1985.Spika, J.S., N. Schaffer, N. Hargrett-Bean, et al. Risk fac<strong>to</strong>rs for infant botulism in theUnited States. Am J Dis Child 143:828–832, 1989.Suen, J.C., C.L. Hatheway, A.G. Steigerwalt, D.J. Brenner. Genetic confirmation of identitiesof neuro<strong>to</strong>xigenic Clostridium baratii <strong>and</strong> Clostridium butyricum implicated as agents ofinfant botulism. J Clin Microbiol 26:2191–2192, 1988a.Suen, J.C., C.L. Hatheway, A.G. Steigerwalt, D.J. Brenner. Clostridium argentinense spnov; a genetically homogenous group of all strains of Clostridium botulinum <strong>to</strong>xin type G <strong>and</strong>some non<strong>to</strong>xigenic strains previously identified as Clostridium subterminale <strong>and</strong> Clostridiumhastiforme. Int J Syst Bacteriol 38:375–381, 1988b.Swerczek, T.W. Toxicoinfectious botulism in foals <strong>and</strong> adult horses. J Am Vet Med Assoc176:217–220, 1980.Terranova, W., J.G. Breman, R.P. Locey, S. Speck. Botulism type B: Epidemiologic aspectsof an extensive outbreak. Am J Epidemiol 180:150–156, 1978.Thomas, R.J., D.V. Rosenthal, R.J. Rogers. A Clostridium botulinum type B vaccine forprevention of shaker foal syndrome. Aust Vet J 65:78–80, 1988.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Botulism in the United States 1899–1973. H<strong>and</strong>book forEpidemiologists, Clinicians, <strong>and</strong> Labora<strong>to</strong>ry Workers. Atlanta: CDC; 1974.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Wound botulism associated with parenteral cocaine abuse.MMWR Morb Mortal Wkly Rep 31:87–88, 1982.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). International outbreak of type E botulism associated withungutted, salted whitefish. MMWR Morb Mortal Wkly Rep 36:812–813, 1987.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Outbreak of type E botulism associated with an uneviscerated,salt-cured fish product—New Jersey, 1992. MMWR Morb Mortal Wkly Rep 41:521–522, 1992.Ying, S., C. Shuan. Botulism in China. Rev Infec Dis 8:984–990, 1986.BRUCELLOSISICD-10 A23.0 brucellosis due <strong>to</strong> Brucella melitensis; A23.1 brucellosisdue <strong>to</strong> Brucella abortus; A23.2 brucellosis due <strong>to</strong> Brucella suis;A23.3 brucellosis due <strong>to</strong> Brucella canisSynonyms: Meli<strong>to</strong>coccosis, undulant fever, Malta fever, Mediterranean fever (inman); contagious abortion, infectious abortion, epizootic abortion (in animals);Bang’s disease (in cattle).Etiology: Six species are presently known in the genus Brucella: B. melitensis, B.abortus, B. suis, B. neo<strong>to</strong>mae, B. ovis, <strong>and</strong> B. canis.The first three species (called “classic brucella”) have been subdivided in<strong>to</strong> biovarsthat are distinguished by their different biochemical characteristics <strong>and</strong>/or reac-


BRUCELLOSIS 41tions <strong>to</strong> the monospecific A. (abortus) <strong>and</strong> M. (melitensis) sera. Thus, B. melitensisis subdivided in<strong>to</strong> three biovars (1–3); B. abortus, in<strong>to</strong> seven (1–7)—biovars 7 <strong>and</strong>8 were discarded <strong>and</strong> the current biovar 7 corresponds <strong>to</strong> 9 in the old classification;<strong>and</strong> B. suis, in<strong>to</strong> five (1–5). From an epidemiological viewpoint, the taxonomicsystem of the genus Brucella has eliminated confusion arising from the naming ofnew species or subspecies that did not agree with epidemiological reality. Moreover,typing by biovars constitutes a useful research <strong>to</strong>ol in that field. The characteristicsof B. abortus determined by conventional methods vary greatly, such as sensitivityor <strong>to</strong>lerance <strong>to</strong> aniline dyes, production of H 2S, <strong>and</strong> CO 2requirements for growth.Less plasticity is shown by B. melitensis or B. suis (Meyer, 1984). In various partsof the world, strains of B. abortus <strong>and</strong>, <strong>to</strong> a lesser extent, of B. suis or B. melitensishave been discovered that are difficult <strong>to</strong> place within the current scheme, in thatthey differ in some characteristics (Ewalt <strong>and</strong> Forbes, 1987; Corbel et al., 1984;Banai et al., 1990).However, the genome of the genus Brucella is very homogeneous as shown byVerger et al. (1985) in a DNA:DNA hybridization study. These researchers proposemaintaining a single species, B. melitensis, subdivided in<strong>to</strong> six biogroups, whichwould correspond <strong>to</strong> the six previous species. For all practical purposes, <strong>and</strong> especiallyfor epidemiological purposes, the previous scheme that divides the genus in<strong>to</strong>species <strong>and</strong> biovars is still in effect.Geographic Distribution: Worldwide. The distribution of the different species ofBrucella <strong>and</strong> their biovars varies with geographic areas. B. abortus is the most widespread;B. melitensis <strong>and</strong> B. suis are irregularly distributed; B. neo<strong>to</strong>mae was isolatedfrom desert rats (Neo<strong>to</strong>ma lepida) in Utah (USA), <strong>and</strong> its distribution is limited<strong>to</strong> natural foci, as the infection has never been confirmed in man or domesticanimals. Infection by B. canis has been confirmed in many countries on several continents,<strong>and</strong> its worldwide distribution can be asserted. B. ovis seems <strong>to</strong> be found inall countries where sheep raising is an important activity.Occurrence in Man: Each year about a half million cases of brucellosis occur inhumans around the world (<strong>WHO</strong>, 1975). The prevalence of the infection in animalreservoirs provides a key <strong>to</strong> its occurrence in humans. B. abortus <strong>and</strong> B. suis infectionsusually affect occupational groups, while B. melitensis infections occur morefrequently than the other types in the general population. The greatest prevalence inman is found in those countries with a high incidence of B. melitensis infectionamong goats, sheep, or both species. The Latin American countries with the largestnumber of recorded cases are Argentina, Mexico, <strong>and</strong> Peru. The same pattern holdstrue for Mediterranean countries, Iran, the former Soviet Union, <strong>and</strong> Mongolia.In Saudi Arabia, 7,893 human cases of brucellosis were recorded in 1987 (74 per100,000 inhabitants). Brucellosis probably became very important in public healthbecause during the period 1979 <strong>to</strong> 1987, Saudi Arabia imported more than 8 millionsheep, more than 2 million goats, more than 250,000 cattle, <strong>and</strong> other animals (buffalo,camels). In Iran, 71,051 cases (13 per 100,000) were recorded in 1988 <strong>and</strong> it isestimated that 80,000 cases have occurred each year since 1989. In Turkey, 5,003cases (9 per 100,000) were recorded in 1990, an incidence three times higher th<strong>and</strong>uring the period 1986–1989 (3 per 100,000).Programs for the control <strong>and</strong> eradication of bovine brucellosis markedly reducethe incidence of disease in humans. For example, in the United States, 6,321 cases


42 BACTERIOSESwere recorded in 1947, while in the period 1972–1981, the annual average was 224cases (CDC, 1982). In Denmark, where some 500 cases per year were reportedbetween 1931 <strong>and</strong> 1939, human brucellosis had disappeared by 1962 as a result ofthe eradication of the infection in animals. In Uruguay, where there is no animalreservoir of B. melitensis <strong>and</strong> where the few foci of B. suis had been eliminated(although they have recently been reintroduced through importation), the disease inhumans has almost disappeared since compulsory vaccination of calves was begunin 1964. China <strong>and</strong> Israel have been able <strong>to</strong> significantly reduce the incidence ofhuman brucellosis thanks <strong>to</strong> vaccination campaigns for sheep <strong>and</strong> goats. In the westernMediterranean area, there has also been a marked reduction of human brucellosiscases caused by B. melitensis due <strong>to</strong> vaccination of the small ruminants withthe Rev. 1 vaccine. In Spain, for example, the incidence fell from 4,683 cases in1988 <strong>to</strong> 3,041 in 1990.Occurrence in Animals: Bovine brucellosis is found worldwide, but it has beeneradicated in Finl<strong>and</strong>, Norway, Sweden, Denmark, the Netherl<strong>and</strong>s, Belgium,Switzerl<strong>and</strong>, Germany, Austria, Hungary, the former Czechoslovakia, Rumania, <strong>and</strong>Bulgaria, as well as other countries (Timm, 1982; Kasyanov <strong>and</strong> Aslanyan, 1982).Most European countries are free of bovine brucellosis (García-Carrillo <strong>and</strong> Lucero,1993). The large meat-producing countries, such as France, Great Britain, Australia,New Zeal<strong>and</strong>, Canada, <strong>and</strong> the United States, among others, are free of bovine brucellosisor close <strong>to</strong> being so. Three important cattle-raising countries, Argentina,Brazil, <strong>and</strong> Mexico, still have limited control programs. A country-by-countryanalysis can be found in a monograph on bovine brucellosis (García-Carrillo <strong>and</strong>Lucero, 1993). In the rest of the world, rates of infection vary greatly from one country<strong>to</strong> another <strong>and</strong> between regions within a country. The highest prevalence is seenin dairy cattle. In many countries, including most of those Latin American countriesthat have no control programs, the data are unreliable. Nevertheless, available informationindicates that it is one of the most serious <strong>diseases</strong> in cattle in Latin Americaas well as in other developing areas. Official estimates put annual losses from bovinebrucellosis in Latin America at approximately US$ 600 million, which explains thepriority given by animal health services <strong>to</strong> control of this disease.Swine brucellosis is infrequent <strong>and</strong> occurs sporadically in most of Europe, Asia,<strong>and</strong> Oceania. In China, B. suis biovar 3 was introduced with breeding s<strong>to</strong>ck fromHong Kong in 1954 <strong>and</strong> spread rapidly through the southern part of the country (Lu<strong>and</strong> Zhang, 1989). In many European countries, swine brucellosis shows an epidemiologicalrelationship <strong>to</strong> brucellosis caused by B. suis biovar 2 in hares (Lepuseuropaeus). With the new swine-breeding technology, swine have little access <strong>to</strong>hares <strong>and</strong> outbreaks have thus shown a marked decline. The disease has never beenpresent in Finl<strong>and</strong>, Norway, Great Britain, <strong>and</strong> Canada. Many predominantlyMuslim countries <strong>and</strong> Israel are probably free of B. suis infection as a result of religiousbeliefs that have limited swine raising (Timm, 1982).In most of Latin America, swine brucellosis is enzootic <strong>and</strong>, while the availabledata have little statistical value, this region is thought <strong>to</strong> have the highest prevalencein the world. However, recent surveys of breeding operations for purebreds <strong>and</strong>hybrids in Argentina <strong>and</strong> Rio Gr<strong>and</strong>e do Sul (Brazil) have shown the percentage ofinfected herds <strong>to</strong> be low. The problem is possibly rooted in commercial operationswhere animals of different origins are brought <strong>to</strong>gether. Thus far, only B. suis bio-


BRUCELLOSIS 43var 1, which predominates worldwide, has been confirmed from Latin America.Biovar 2 is limited <strong>to</strong> pigs <strong>and</strong> hares in central <strong>and</strong> western Europe, while biovar 3is limited <strong>to</strong> the corn belt of the United States <strong>and</strong> <strong>to</strong> some areas of Asia <strong>and</strong> Africa.The US <strong>and</strong> Cuba have successful national eradication programs.Goat <strong>and</strong> sheep brucellosis constitute a significant problem in the Mediterraneanbasin of Europe <strong>and</strong> Africa, in the southeastern part of the former Soviet Union, inMongolia, in the Middle East, <strong>and</strong> Saudi Arabia. In Latin America, the prevalenceof B. melitensis infection in goats is high in Argentina, Mexico, <strong>and</strong> Peru. To date,sheep infection with B. melitensis in Argentina has been identified only in flocks livingwith infected goats in the north of the country (Ossola <strong>and</strong> Szyfres, 1963). InVenezuela’s goat-raising region, a serological <strong>and</strong> bacteriological examination wasconducted in 1987. B. abortus biovar 1 was isolated from milk <strong>and</strong> lymph nodes. B.melitensis was not isolated (De Lord et al., 1987). Goat brucellosis does not appear<strong>to</strong> exist in Brazil, which has a sizable number of goats. In Chile, where the rate ofinfection in Cajón de Maipo was significant, the Government reported that the diseasehad been eradicated (Chile, Ministerio de Agricultura, 1987). Other Americancountries, including the US, are free of goat brucellosis at the present time.Ram epididymitis caused by B. ovis is widespread. It has been confirmed in NewZeal<strong>and</strong>, Australia, Africa, <strong>and</strong> Europe. It is present in Argentina, Brazil (RioGr<strong>and</strong>e do Sul), Chile, Peru, Uruguay, <strong>and</strong> the US, i.e., in all American countrieswhere sheep are raised on a large scale. Prevalence is high.Infection of dogs with B. canis has been found in almost every country in theworld where it has been studied. Prevalence varies according <strong>to</strong> region <strong>and</strong> diagnosticmethod used. It constitutes a problem for some dog breeders, since it causesabortions <strong>and</strong> infertility, but the infection is also found in family dogs <strong>and</strong> strays. Inthe latter, the incidence of infection is usually higher. In a study carried out inMexico City, for example, 12% of 59 stray dogs were positive in the isolation of theetiologic agent (Flores-Castro et al., 1977).The Disease in Man: Man is susceptible <strong>to</strong> infection caused by B. melitensis, B.suis, B. abortus, <strong>and</strong> B. canis. No human cases caused by B. ovis, B. neo<strong>to</strong>mae, or B.suis biovar 2 have been confirmed. The most pathogenic <strong>and</strong> invasive species for manis B. melitensis, followed in descending order by B. suis, B. abortus, <strong>and</strong> B. canis.In general, the incubation period is one <strong>to</strong> three weeks, but may sometimes be severalmonths. The disease is septicemic, with sudden or insidious onset, <strong>and</strong> isaccompanied by continued, intermittent, or irregular fever. The symp<strong>to</strong>ma<strong>to</strong>logy ofacute brucellosis, like that of many other febrile <strong>diseases</strong>, includes chills <strong>and</strong> profusesweating. Weakness is an almost constant symp<strong>to</strong>m, <strong>and</strong> any exercise producespronounced fatigue. Temperature can vary from normal in the morning <strong>to</strong> 40°C inthe afternoon. Sweating characterized by a peculiar odor occurs at night. Commonsymp<strong>to</strong>ms are insomnia, sexual impotence, constipation, anorexia, headache,arthralgia, <strong>and</strong> general malaise. The disease has a marked effect on the nervous system,evidenced by irritation, nervousness, <strong>and</strong> depression. Many patients haveenlarged peripheral lymph nodes or splenomegaly <strong>and</strong> often hepa<strong>to</strong>megaly, butrarely jaundice. Hepa<strong>to</strong>megaly or hepa<strong>to</strong>splenomegaly is particularly frequent inpatients infected by B. melitensis (Pfischner et al., 1957). Brucella organisms localizeintracellularly in tissues of the reticuloendothelial system, such as lymph nodes,bone marrow, spleen, <strong>and</strong> liver. Tissue reaction is granuloma<strong>to</strong>us. The duration of


44 BACTERIOSESthe disease can vary from a few weeks or months <strong>to</strong> several years. Modern therapyhas considerably reduced the disease’s duration as well as the incidence of relapses.At times, it produces serious complications, such as encephalitis, meningitis, peripheralneuritis, spondylitis, suppurative arthritis, vegetative endocarditis, orchitis, seminalvesiculitis, <strong>and</strong> prostatitis. A chronic form of the disease occurs in some patients<strong>and</strong> may last many years, with or without the presence of localized foci of infection.The symp<strong>to</strong>ms are associated with hypersensitivity. Diagnosis of chronic brucellosisis difficult.Separate mention should be made of human infection caused by the B. abortusstrain 19 vaccine, which is the vaccine used most often <strong>to</strong> protect cattle. Cases havebeen described of accidents among those administering the vaccine (veterinarians<strong>and</strong> assistants) who have pricked a finger or h<strong>and</strong> with the syringe needle or havegotten aerosol in their eyes. If someone has no prior exposure <strong>to</strong> brucellae <strong>and</strong> hasno antibodies <strong>to</strong> the agent, the disease sets in abruptly after a period of 8 <strong>to</strong> 30 days.The course of the disease is usually shorter <strong>and</strong> more benign than that caused by thefield strains of B. abortus, but there are severe cases that require hospitalization. Inindividuals who have been exposed <strong>to</strong> brucellae, as is usually the case with veterinarians<strong>and</strong> vaccina<strong>to</strong>rs, a different, allergic-type syndrome appears that is characterizedby painful swelling at the inoculation site. After some hours, the patient mayexperience systemic symp<strong>to</strong>ms similar <strong>to</strong> those described in individuals infected bystrain 19 without prior exposure. The symp<strong>to</strong>ms usually abate in a few days with orwithout treatment. Local <strong>and</strong> general symp<strong>to</strong>ms may recur if the person has anotheraccident (Young, 1989). Considering the millions of doses of strain 19 vaccine usedeach year throughout the world, the rate of incidence of the disease due <strong>to</strong> this strainis insignificant.Another strain that is used <strong>to</strong> vaccinate small ruminants, B. melitensis Rev. 1, canalso infect the vaccina<strong>to</strong>r. Under the aegis of the World Health Organization (<strong>WHO</strong>)<strong>and</strong> its collaborative centers, Rev. 1 vaccine was administered <strong>to</strong> 6 million animalsin Mongolia between 1974 <strong>and</strong> 1977. Six trained vaccina<strong>to</strong>rs inoculated themselvesaccidentally; four of them showed clinical symp<strong>to</strong>ms but recovered after immediatehospital treatment.There are many infections that occur asymp<strong>to</strong>matically in areas with enzooticbrucellosis, particularly the bovine form.The recommended treatment for acute brucellosis is a daily dose of 600 mg <strong>to</strong> 900mg of rifampicin, combined with 200 mg per day of doxycycline for at least sixweeks. Relapses are very rare with this treatment. If there is a Jarisch-Herxheimerreaction upon starting antibiotic treatment, intravenous administration of cortisol isrecommended. Sometimes various series of treatment are needed. If antibiotic therapyis not successful, a chronic focus of infection should be sought, particularly ininfections caused by B. melitensis <strong>and</strong> B. suis (<strong>WHO</strong>, 1986). In the event of arelapse, the treatment indicated above should be restarted. Steroids may be administered<strong>to</strong> counteract <strong>to</strong>xicity in patients who are very ill (Benenson, 1992).The Disease in Animals: The principal symp<strong>to</strong>m in all animal species is abortionor premature expulsion of the fetus.CATTLE: The main pathogen is B. abortus. Biovar 1 is universal <strong>and</strong> predominantamong the seven that occur in the world. The distribution of the different biovarsvaries geographically. In Latin America, biovars 1, 2, 3, 4, <strong>and</strong> 6 have been con-


BRUCELLOSIS 45firmed, with biovar 1 accounting for more than 80% of the isolations. In the UnitedStates, biovars 1, 2, <strong>and</strong> 4 have been isolated. In eastern Africa <strong>and</strong> China, biovar 3predominates <strong>and</strong> affects both native cattle <strong>and</strong> buffalo (Timm, 1982). Biovar 5,which occurred in cattle in Great Britain <strong>and</strong> Germany, has biochemical <strong>and</strong> serologicalcharacteristics similar <strong>to</strong> B. melitensis. This similarity was a source of confusionfor years until new methods of species identification (oxidative metabolism<strong>and</strong> phagocy<strong>to</strong>lysis) established the biovar as B. abortus. The other biovars also havea more or less marked geographic distribution. Cattle can also become infected byB. suis <strong>and</strong> B. melitensis when they share pasture or facilities with infected pigs,goats, or sheep. The infection in cattle caused by heterologous species of Brucellaare usually more transient than that caused by B. abortus. However, such crossinfectionsare a serious public health threat, since these brucellae, which are highlypathogenic for man, can pass in<strong>to</strong> cow’s milk. Infection caused by B. suis is not very<strong>common</strong>. By contrast, infections caused by B. melitensis have been seen in severalcountries, with a course similar <strong>to</strong> those caused by B. abortus.In natural infections, it is difficult <strong>to</strong> measure the incubation period (from time ofinfection <strong>to</strong> abortion or premature birth), since it is not possible <strong>to</strong> determine themoment of infection. Experiments have shown that the incubation period varies considerably<strong>and</strong> is inversely proportional <strong>to</strong> fetal development: the more advanced thepregnancy, the shorter the incubation period. If the female is infected orally duringthe breeding period, the incubation period can last some 200 days, while if she isexposed six months after being bred, incubation time is approximately two months.The period of “serologic incubation” (from the time of infection <strong>to</strong> the appearanceof antibodies) lasts several weeks <strong>to</strong> several months. The incubation period variesaccording <strong>to</strong> such fac<strong>to</strong>rs as the virulence <strong>and</strong> dose of bacteria, the route of infection,<strong>and</strong> the susceptibility of the animal.The predominant symp<strong>to</strong>m in pregnant females is abortion or premature or fulltermbirth of dead or weak calves. In general, abortion occurs during the second halfof the pregnancy, often with retention of the placenta <strong>and</strong> resultant metritis, whichmay cause permanent infertility. It is estimated that the infection causes a 20% <strong>to</strong>25% loss in milk production as a result of interrupted lactation due <strong>to</strong> abortion <strong>and</strong>delayed conception. Cows artificially inseminated with infected semen may comein<strong>to</strong> estrus repeatedly, as happens in cases of vibriosis or trichomoniasis.Nonpregnant females show no clinical symp<strong>to</strong>ms <strong>and</strong>, if infected prior <strong>to</strong> breeding,often do not abort.In bulls, brucellae may become localized in the testicles <strong>and</strong> adjacent genitalgl<strong>and</strong>s. When the clinical disease is evident, one or both testicles may becomeenlarged, with decreased libido <strong>and</strong> infertility. Sometimes a testicle may atrophy due<strong>to</strong> adhesions <strong>and</strong> fibrosis. Seminal vesiculitis <strong>and</strong> ampullitis are <strong>common</strong>.Occasionally, hygromas <strong>and</strong> arthritis are observed in cattle.Brucellae entering the animal’s body multiply first in the regional lymph nodes<strong>and</strong> are later carried by the lymph <strong>and</strong> blood <strong>to</strong> different organs. Some two weeksafter experimental infection, bacteremia can be detected <strong>and</strong> it is possible <strong>to</strong> isolatethe agent from the bloodstream. Brucella organisms are most <strong>common</strong>ly found inthe lymph nodes, uterus, udder, spleen, liver, <strong>and</strong>, in bulls, the genital organs. Largequantities of erythri<strong>to</strong>l, a carbohydrate that stimulates the multiplication of brucellae,have been found in cow placentas. This could explain the high susceptibility ofbovine fetal tissues.


46 BACTERIOSESOnce an infected cow aborts or gives birth normally, the pathogen does not remainlong in the uterus. The infection becomes chronic <strong>and</strong> the brucellae are harbored inthe cow’s lymph nodes <strong>and</strong> mammary gl<strong>and</strong>s. Brucellae may remain in the udder foryears (García-Carrillo <strong>and</strong> Lucero, 1993).Individual animals within a herd manifest different degrees of susceptibility <strong>to</strong>infection depending on their age <strong>and</strong> sex. Male <strong>and</strong> female calves up <strong>to</strong> 6 months ofage are not very susceptible <strong>and</strong> generally experience only transi<strong>to</strong>ry infections. Abull calf fed milk containing brucella organisms can harbor the agent in its lymphnodes, but after six <strong>to</strong> eight weeks without ingesting the contaminated food, the animalusually rids itself of the infection.Heifers kept separate from cows, as is routine in herd management, often havelower infection rates than adult cows. Heifers exposed <strong>to</strong> infection before breedingcan become infected, but generally do not abort. In view of this, at the beginning ofthe century heifers were inoculated before breeding with virulent strains or withstrains of unknown virulence <strong>to</strong> prevent abortion. This practice had <strong>to</strong> be ab<strong>and</strong>oned,however, when it was found that a large number of animals remained infected.Cows, especially when pregnant, are the most susceptible; infection is <strong>common</strong><strong>and</strong> abortion frequently results.Bulls are also susceptible, although some researchers maintain that they are moreresistant <strong>to</strong> infection than females. This conclusion may reflect herd managementpractices more than natural resistance in males, however, since bulls are usually keptseparate from cows. On the other h<strong>and</strong>, neutered males <strong>and</strong> females do not play arole in the epizootiology of brucellosis, since they cannot transmit brucellae <strong>to</strong> theexterior environment.In addition <strong>to</strong> age <strong>and</strong> sex, it is important <strong>to</strong> take individual susceptibility in<strong>to</strong>account. Even in the most susceptible categories—cows <strong>and</strong> heifers—some animalsnever become infected, or if they do, their infection is transient. Some less susceptiblecows have generalized infections <strong>and</strong> suffer losses in reproductive function <strong>and</strong>milk production for one or more years, but then gradually recover. In such animals,the agglutination titer becomes negative, the shedding of brucellae may cease, <strong>and</strong>both reproductive function <strong>and</strong> milk production return <strong>to</strong> normal. However, mostcows become infected, <strong>and</strong> their agglutination titers remain positive for many yearsor for life; although after one or two abortions they may give birth normally <strong>and</strong>resume normal production of milk, many continue <strong>to</strong> carry <strong>and</strong> shed brucellae. Othercows remain <strong>to</strong>tally useless for breeding <strong>and</strong> milk production.In a previously uninfected herd, brucellosis spreads rapidly from animal <strong>to</strong> animal,<strong>and</strong> for one or two years there are extensive losses from abortions, infertility,decreased milk production, <strong>and</strong> secondary genital infections. This acute or activephase of the disease is characterized by a large number of abortions <strong>and</strong> a high rateof reac<strong>to</strong>rs in serological tests. Because of individual differences in susceptibility <strong>to</strong>the infection, not all animals become infected <strong>and</strong> not all those that are serologicallypositive abort. After a year or two, the situation stabilizes <strong>and</strong> the number of abortionsdecreases. It is estimated that only between 10% <strong>and</strong> 25% of the cows willabort a second time. In this stabilization phase, it is primarily the heifers—not previouslyexposed <strong>to</strong> the infection—that become infected <strong>and</strong> may abort. A final,decline phase can be observed in small <strong>and</strong> self-contained herds. In this phase, theinfection rate gradually decreases, <strong>and</strong> most of the cows return <strong>to</strong> normal reproductivefunction <strong>and</strong> milk production. Nevertheless, when a sufficient number of sus-


BRUCELLOSIS 47ceptible animals accumulates—either heifers from the same herd or newly introducedanimals—a second outbreak can occur. In large herds, there are alwaysenough susceptible animals <strong>to</strong> maintain the infection, <strong>and</strong> abortions continue.Trading <strong>and</strong> movement of animals also help maintain active infection.SWINE: The main etiologic agent of brucellosis in swine is B. suis. In LatinAmerica, only biovar 1 infection has been confirmed, while in the United States both1 <strong>and</strong> 3 have been involved. Biovar 2 is found only in Europe. Infection by biovars1 <strong>and</strong> 3 is spread directly or indirectly from pig <strong>to</strong> pig. In contrast, biovar 2 (orDanish biovar) is transmitted <strong>to</strong> pigs when they ingest European hares (Lepuseuropaeus). Pigs can also be infected by B. abortus, although it is less pathogenicfor pigs <strong>and</strong> apparently not transmitted from one animal <strong>to</strong> another; the infection isgenerally asymp<strong>to</strong>matic, with the affected organisms limited <strong>to</strong> the lymph nodes ofthe head <strong>and</strong> neck.When brucellosis is introduced in<strong>to</strong> a previously healthy herd, the symp<strong>to</strong>ms arethose of acute disease: abortions, infertility, birth of weak piglets, orchitis, epididymitis,<strong>and</strong> arthritis. In small herds, the infection tends <strong>to</strong> die out or decrease inseverity because of a lack of susceptible animals owing <strong>to</strong> the normal sale of somepigs <strong>and</strong> <strong>to</strong> the spontaneous recovery of others. In large herds, the infection is persistent<strong>and</strong> transmitted from one generation <strong>to</strong> the next.Early abortions, which occur when the female is infected during coitus, generallygo unnoticed under free-range conditions. The aborted fetuses are eaten by the pigs,<strong>and</strong> the only abnormality that may be noted by the owner is the sows’ repeatedestrus. Abortions occur in the second half of gestation when the females are infectedafter one or two months of pregnancy. Affected sows rarely have a second abortion,<strong>and</strong> females infected before sexual maturity rarely abort.Infection is usually temporary in suckling pigs. However, a few may retain theinfection <strong>and</strong> become carriers. It rarely results in recognizable clinical symp<strong>to</strong>ms.Occasionally, arthritis is observed, but transient bacteremia <strong>and</strong> low agglutinationtiters may be found.In infected pigs, abscesses of different sizes frequently occur in organs <strong>and</strong> tissues.Spondylitis is often found.Infection of the genital organs lasts for a shorter period of time in the female thanin the male. In the latter it may last for the life of the animal.GOATS: The main etiologic agent of brucellosis in goats is B. melitensis with itsthree biovars. All types of goats are susceptible <strong>to</strong> infection by B. melitensis.Infection by B. suis <strong>and</strong> B. abortus has occasionally been found.The symp<strong>to</strong>ma<strong>to</strong>logy is similar <strong>to</strong> that observed in other species of animals <strong>and</strong> themain symp<strong>to</strong>m is abortion, which occurs most frequently in the third or fourth monthof pregnancy. In natural infections occurring in the field, other symp<strong>to</strong>ms, such asarthritis, mastitis, spondylitis, <strong>and</strong> orchitis, are rarely found. These symp<strong>to</strong>ms can beseen when the animals are inoculated experimentally with large doses of the agent.Sexually mature female goats that are not pregnant are susceptible <strong>and</strong> suffer from achronic infection that may have no clinical symp<strong>to</strong>ms, but that represents a risk forthe other animals in the flock. Infection of the mammary gl<strong>and</strong> is <strong>common</strong> (Al<strong>to</strong>n,1985). In chronically infected flocks, the signs of the disease are generally not veryapparent. Gross pathological lesions are also not usually evident, though thepathogen can frequently be isolated from a large number of tissues <strong>and</strong> organs.


48 BACTERIOSESSeveral researchers have observed that young goats can be born with the infectionor become infected shortly after birth. Most of them recover spontaneously beforereaching reproductive age, but in some the infection may persist longer.The primitive conditions under which goats are raised constitute one of the mostimportant fac<strong>to</strong>rs in the maintenance <strong>and</strong> spread of the infection in Latin America(Argentina, Mexico, <strong>and</strong> Peru) <strong>and</strong> in the rest of the developing world. In goat-raisingareas, it is <strong>common</strong> <strong>to</strong> find community-shared pastures, a lack of hygiene in makeshiftcorrals, nomadic flocks, <strong>and</strong> owners with little underst<strong>and</strong>ing of herd management.SHEEP: Two disease entities are distinguishable in sheep: classic brucellosis <strong>and</strong>ram epididymitis. Classic brucellosis is caused by B. melitensis <strong>and</strong> constitutes apublic health problem equally or even more important than goat brucellosis in areaswhere the agent is found outside the American continent. In Latin America, theinfection in sheep has been confirmed only in some mixed goat <strong>and</strong> sheep flocksraised far away from intensive sheep-raising areas.While sheep brucellosis is similar in its symp<strong>to</strong>ma<strong>to</strong>logy <strong>to</strong> the disease in goats,sheep appear <strong>to</strong> be more resistant <strong>to</strong> infection <strong>and</strong>, in mixed flocks, fewer sheep thangoats are found <strong>to</strong> be infected. Susceptibility varies from breed <strong>to</strong> breed. Maltesesheep are very resistant, while Middle Eastern Awassi (fat tail) sheep are very susceptible(Al<strong>to</strong>n, 1985). Abortions are also less <strong>common</strong>. The infection tends <strong>to</strong> disappearspontaneously, <strong>and</strong> the high prevalence of the disease in some areas can bestbe attributed <strong>to</strong> poor herd management.Occasionally, sheep have been found <strong>to</strong> be infected by B. suis (biovar 2 inGermany) <strong>and</strong> B. abortus (in various parts of the world). These agents are not verypathogenic for sheep; they are acquired through contact with infected animals ofother species, <strong>and</strong> are usually not transmitted from sheep <strong>to</strong> sheep. However, transmissioncan occur, as in the case described in an outbreak occurring on a ranch inthe US (Luchsinger <strong>and</strong> Anderson, 1979).Ram epididymitis is caused by B. ovis. The clinical signs consist of genital lesionsin rams, associated with varying degrees of sterility. Sometimes the infection inpregnant ewes can cause abortion or neonatal mortality. Epididymitis is generallyunilateral but can be bilateral <strong>and</strong> the tail of the organ is most <strong>common</strong>ly affected.Adhesions may occur in the tunica vaginalis testis, <strong>and</strong> the testicle may be atrophiedwith varying degrees of fibrosis. Lesions cannot be seen or palpated in manyinfected rams, even though B. ovis may be isolated from their semen. Some of theseanimals develop lesions in more advanced stages of the disease. Early in the infection,the semen contains many brucellae, but with time the number decreases, <strong>and</strong>eventually the semen may be free of the infectious agent. When localized in the kidneys,B. ovis is also shed through the urine.HORSES: B. abortus <strong>and</strong> B. suis have been isolated from this species. The diseaseusually manifests itself in the form of fistulous bursitis, “poll evil” <strong>and</strong> “fistulouswithers.” Abortions are rare but they do occur (Robertson et al., 1973). B. abortushas been isolated from horse feces, but this is un<strong>common</strong>. Horses acquire the infectionfrom cattle or swine, but transmission from horses <strong>to</strong> cattle has also beenproven. Man can contract the infection from horses that have open lesions. In general,horses are more resistant <strong>to</strong> the infection. Cases of horse-<strong>to</strong>-horse transmissionare unknown. In areas where there is a high rate of infection, it is <strong>common</strong> <strong>to</strong> findhorses with high agglutination titers.


BRUCELLOSIS 49DOGS AND CATS: Sporadic cases of brucellosis caused by B. abortus, B. suis, <strong>and</strong>B. melitensis occur in dogs. They acquire the infection primarily by eating contaminatedmaterial, especially fetuses, afterbirth, <strong>and</strong> milk. The course of the infectionis usually subclinical, but sometimes the symp<strong>to</strong>ma<strong>to</strong>logy can be severe, with fever,emaciation, orchitis, anestrus, arthritis, <strong>and</strong> sometimes abortion. Cases of dog-<strong>to</strong>dogtransmission are rare. In some cases, the infection may last for more than 150days. Although it is rare, dogs can eliminate brucellae in their urine, vaginal secretions,feces, <strong>and</strong> aborted fetuses. A study conducted in Canada collected 14 dogsfrom 10 cattle properties with bovine brucellosis. Positive cultures were obtainedfrom vaginal mucus <strong>and</strong> from the bladder of a single dog. The final positive vaginalsecretion sample was obtained 464 days after the probable date when the dog wasinfected. In other dogs, Brucella was isolated from organs that do not discharge <strong>to</strong>the environment (Forbes, 1990). Several human cases have been described in whichthe source of infection was dogs (especially fetuses).A canine disease that occurs worldwide <strong>and</strong> can reach epizootic proportions isthat caused by B. canis. This form of brucellosis is characterized by a prolongedafebrile bacteremia, embryonic death, abortions, prostatitis, epididymitis, scrotaldermatitis, lymphadenitis, <strong>and</strong> splenitis. Abortion occurs about 50 days in<strong>to</strong> gestation.The pups may be stillborn at full term or die a few days after birth. Survivorsusually have enlarged lymph nodes <strong>and</strong> often have bacteremia.In an experimental treatment, minocycline (27.5 mg/kg twice a day) was administered<strong>to</strong> 18 infected dogs. Fifteen of them had positive cultures in au<strong>to</strong>psies conductedbetween 6 <strong>and</strong> 28 weeks after treatment ended (<strong>WHO</strong>, 1986).Man is susceptible <strong>to</strong> B. canis, though less so than <strong>to</strong> classic brucellae. Severalcases have been confirmed in the United States, Mexico, Brazil, <strong>and</strong> Argentina inlabora<strong>to</strong>ry <strong>and</strong> kennel personnel as well as in members of families with infecteddogs.Cats are resistant <strong>to</strong> Brucella <strong>and</strong> no cases of natural disease occurrence are known.OTHER DOMESTIC MAMMALS: Brucellosis caused by B. abortus occurs in domesticbuffalo (Bubalus bubalis) <strong>and</strong> in yaks (Bos grunniens) with symp<strong>to</strong>ma<strong>to</strong>logysimilar <strong>to</strong> that in cattle. The disease has also been observed in Old World camels(Camelus bactrianus), in dromedaries (Camelus dromedarius), <strong>and</strong> in AmericanCamelidae. Infection in Camelidae is caused primarily by B. melitensis, although B.abortus has been isolated (Al-Khalaf <strong>and</strong> El-Khaladi, 1989). An outbreak of brucellosiscaused by B. mellitus biovar 1, accompanied by abortions <strong>and</strong> neonatal mortality,occurred on an alpaca (Lama pacos) ranch in the high plateau (altiplano)region of Peru; a serious outbreak also occurred in the human population of thatranch (Acosta et al., 1972).WILD ANIMALS: Natural infections caused by Brucella occur in a wide range ofwild species. There are natural foci of infection, for example, among the desert ratsof the United States (Neo<strong>to</strong>ma lepida), which are the reservoir of B. neo<strong>to</strong>mae. InKenya, B. suis biovar 3 has been isolated from two species of rodents (Arvicanthisniloticus <strong>and</strong> Mas<strong>to</strong>mys natalensis). In Australia, there are as yet unclassified biovarsof Brucella in various species of rodents. In the Caucasus, rodents infected byBrucella were found; it was initially classified as B. muris <strong>and</strong> later as B. suis biovar5. In Europe, the infection of hares (Lepus europaeus), which are the reservoirof B. suis biovar 2, is transmitted <strong>to</strong> domestic swine. Caribou (Rangifer caribou),


50 BACTERIOSESwhich is the reservoir of B. suis biovar 4 in Alaska, can transmit the infection <strong>to</strong> man<strong>and</strong> <strong>to</strong> sled dogs. The infection can also be transmitted in the opposite direction,from domestic animals <strong>to</strong> wild animals. This is the case in Argentina, where infectionin foxes (Dusicyon gymnocercus, D. griseus) (Szyfres <strong>and</strong> González Tomé,1966) <strong>and</strong> grisons (Galictis furax-huronax) is caused by B. abortus biovar 1, infectionin European hares (Lepus europaeus) is caused by B. suis biovar 1 (Szyfres etal., 1968), <strong>and</strong> that in opossums (Didelphis azarae), by B. abortus biovar 1 <strong>and</strong> B.suis biovar 1 (De la Vega et al., 1979). Carnivores acquire the infection by eatingfetuses <strong>and</strong> afterbirth. There is no evidence that the infection is transmitted from oneindividual <strong>to</strong> another among carnivores, <strong>and</strong> it probably dies out when brucellosis iscontrolled in domestic animals. The situation is different when domestic animalstransmit the infection <strong>to</strong> wild ruminants, such as the steppe antelope (Saiga tatarica)or the American bison (Bison bison), in which brucellosis persists.Fur-bearing animals, such as minks <strong>and</strong> silver foxes, may contract brucellosis whenfed viscera of infected animals, <strong>and</strong> they may in turn transmit this infection <strong>to</strong> man.The etiologic agent has been isolated from many species of arthropods. Ticks canharbor the organism for lengthy periods <strong>and</strong> transmit the infection through biting.They also eliminate the bacteria in their coxal gl<strong>and</strong> secretions. Nevertheless, thenumber of ticks harboring brucellae is insignificant (in one study done in the formerSoviet Union, eight strains of Brucella spp. were isolated from 20,000 ticks) <strong>and</strong>there are few brucellae per tick. The species that have been isolated from arthropodsare B. melitensis <strong>and</strong> B. abortus. In Brazil, B. canis was isolated from specimens ofRhipicephalus sanguineus attached <strong>to</strong> a bitch suffering from brucellosis (Peres et al.,1981). There is consensus that arthropods play only a small role, if any, in the epidemiologyof brucellosis.FOWL: In a few cases, Brucella has been isolated from naturally infected domesticfowl. The symp<strong>to</strong>ma<strong>to</strong>logy described is quite varied, <strong>and</strong> there is no certainty that italways involves brucellosis. The infection may not be evident, with symp<strong>to</strong>ms suchas weight loss, reduction in egg production, <strong>and</strong> diarrhea. Fowl do not play a role inmaintaining the infection in nature. Brucella has been isolated from some wild birdspecies such as ravens (Corvus corvix) <strong>and</strong> crows (Tripanscorax fragilecus).Source of Infection <strong>and</strong> Mode of Transmission: The natural reservoirs of B.abortus, B. suis, <strong>and</strong> B. melitensis are, respectively, cattle, swine, <strong>and</strong> goats <strong>and</strong>sheep. The natural host of B. canis is the dog <strong>and</strong> that of B. ovis is the sheep.INFECTION IN HUMANS: Man is infected by animals through direct contact or indirectlyby ingestion of animal products <strong>and</strong> by inhalation of airborne agents. The relativeimportance of the etiologic agent’s mode of transmission <strong>and</strong> pathway of penetrationvaries with the epidemiological area, the animal reservoirs, <strong>and</strong> theoccupational groups at risk. Fresh cheese <strong>and</strong> raw milk from goats <strong>and</strong> sheepinfected with B. melitensis are the most <strong>common</strong> vehicles of infection <strong>and</strong> can causemultiple cases of human brucellosis. Sometimes more widespread outbreaks occurwhen infected goat’s milk is mixed with cow’s milk. Cow’s milk infected by B.melitensis or B. suis has also been known <strong>to</strong> produce outbreaks of epidemic proportions.Cow’s milk <strong>and</strong> milk products containing B. abortus may give rise <strong>to</strong> sporadiccases. The organisms rarely survive in sour milk, sour cream <strong>and</strong> butter, or fermentedcheese (aged over three months).


BRUCELLOSIS 51In arctic <strong>and</strong> subarctic regions, there have been confirmed cases that resulted fromeating bone marrow or raw meat from reindeer or caribou infected with B. suis biovar4. Brucellae are resistant <strong>to</strong> pickling <strong>and</strong> smoke curing, therefore some meatproducts thus prepared could possibly cause human infection; however, this mode oftransmission has never been verified.It is also possible for raw vegetables <strong>and</strong> water contaminated with the excreta ofinfected animals <strong>to</strong> serve as sources of infection.Transmission by contact predominates in areas where bovine <strong>and</strong> porcine brucellosisare enzootic. Human brucellosis is, for the most part, an occupational diseaseof s<strong>to</strong>ckyard <strong>and</strong> slaughterhouse workers, butchers, <strong>and</strong> veterinarians. The infectionis usually contracted by h<strong>and</strong>ling fetuses <strong>and</strong> afterbirth, or by contact with vaginalsecretions, excreta, <strong>and</strong> carcasses of infected animals. The microorganism entersthrough skin abrasions as well as through the conjunctiva by way of the h<strong>and</strong>s. Inslaughterhouses, prevalence of the disease is higher among recently employed staff.The practice in some companies of employing workers with negative serology ismisguided, since an individual who is asymp<strong>to</strong>matic but has a positive serology isless likely <strong>to</strong> become sick.In areas where goat <strong>and</strong> sheep brucellosis is enzootic, transmission by contact alsooccurs when shepherds h<strong>and</strong>le newborn animals or fetuses. In some countries withhard winters, goats share the beds of goatherds <strong>and</strong> their families for protectionagainst the cold, which results in infection of the whole family (Elberg, 1981).Airborne transmission has been proved by experimentation <strong>and</strong> research. In labora<strong>to</strong>ries,centrifugation of brucellosis suspensions poses a special risk when done incentrifuges that are not hermetically sealed. An epidemic outbreak of 45 casesoccurred among students at Michigan State University (USA) in 1938–1939. The 45students were attending classes on the second <strong>and</strong> third floors of a building thathoused a brucellosis research labora<strong>to</strong>ry in the basement. In the ensuing investigation,it was shown that the only possible means of transmission was by aerosol particles.Subsequent epidemiological studies have supplied proof that airborne transmissionin meat lockers <strong>and</strong> slaughterhouses plays an important role, <strong>and</strong> perhaps ismore frequent than transmission by direct contact with infected tissue. When air inthe killing area is allowed <strong>to</strong> disperse, it leads <strong>to</strong> high rates of infection among workersin adjoining areas. The minimum infective dose for man by way of the respira<strong>to</strong>rypassages seems <strong>to</strong> be small. When the killing area is completely separate, ormaintained at a negative air pressure, the risk <strong>to</strong> surrounding areas is reduced(Kaufmann et al., 1980; Buchanan et al., 1974).Some cases of possible human-<strong>to</strong>-human transmission of brucellosis have beendescribed. One of them occurred in Kuwait due <strong>to</strong> transmission of B. melitensis <strong>to</strong> a30-day-old girl through her mother’s milk. The mother had experienced fever, discomfort,<strong>and</strong> arthralgia for at least two weeks prior <strong>to</strong> the child’s becoming sick. B.melitensis biovar 1 was repeatedly isolated from the blood of both mother <strong>and</strong> child(Lubani et al., 1988). In a hospital labora<strong>to</strong>ry in the US, eight microbiologists wereexposed <strong>to</strong> accidental dispersion of a clinical specimen in aerosol <strong>and</strong> B. melitensisbiovar 3 was isolated from five of them. The spouse of one of the patients becameill six months after her husb<strong>and</strong> had been admitted <strong>to</strong> the hospital <strong>and</strong> B. melitensisof the same biovar was isolated from her blood; it is suspected that the infection wassexually transmitted (Ruben et al., 1991). A probable case of transmission duringchildbirth occurred in Israel. The mother had a fever on the first day postpartum <strong>and</strong>


52 BACTERIOSESFigure 5. Bovine brucellosis (Brucella abortus). Mode of transmission.BullArtificial inseminationCowDirect <strong>and</strong> indirect contact,ingestion of raw milk<strong>and</strong> fresh cheesesCowFetuses, fetalmembranes,vaginal secretionsContact with fetuses,contaminated objectsEnvironmentalcontamination(pasture, forage,water, stable)Ingestion, contactBull <strong>and</strong>CowManB. melitensis biovar 3 was identified in a cervical culture. Cervical <strong>and</strong> blood culturescontinued <strong>to</strong> be positive. Although the child was asymp<strong>to</strong>matic, a positiveblood culture of the same biovar <strong>and</strong> an agglutinating titer of 1:100 were obtained.Splenomegaly was the only abnormality found in the child at 13 days. Prior <strong>to</strong> thesecases, there were descriptions of human-<strong>to</strong>-human transmission due <strong>to</strong> transfusionor bone marrow transplants.INFECTION IN CATTLE (Figure 5): The main sources of infection for cattle arefetuses, afterbirth, <strong>and</strong> vaginal discharges containing large numbers of brucellae. Toa lesser extent, farm areas can be contaminated by fecal matter of calves fed on contaminatedmilk, since not all the organisms are destroyed in the digestive tract.The most <strong>common</strong> route of transmission is the gastrointestinal tract followingingestion of contaminated pasture, feed, fodder, or water. Moreover, cows cus<strong>to</strong>marilylick afterbirth, fetuses, <strong>and</strong> newborn calves, all of which may contain a largenumber of the organisms <strong>and</strong> constitute a very important source of infection. Cows’habit of licking the genital organs of other cows also contributes <strong>to</strong> transmission ofthe infection.It has been shown experimentally that the organism may penetrate broken <strong>and</strong>even intact skin. The extent <strong>to</strong> which this mode of transmission is involved in naturalinfection is unknown.Bang <strong>and</strong> others experimentally reproduced infection <strong>and</strong> disease via the vaginalroute. The results of those experiments indicate that a large number of brucellae arenecessary <strong>to</strong> infect a cow by this means. However, there is no doubt that the


BRUCELLOSIS 53intrauterine route used in artificial insemination is very important in transmitting theinfection. The use of infected bulls for artificial insemination constitutes an importantrisk, since the infection can thus be spread <strong>to</strong> many herds.In closed environments, it is likely that infection is spread by aerosols; airborneinfection has been demonstrated experimentally.Congenital infection <strong>and</strong> the so-called latency phenomenon have also beendescribed. An experiment was carried out in France (Plommet et al., 1973) in whichcalves born <strong>to</strong> cows artificially infected with a high dose of B. abortus were separatedfrom their mothers <strong>and</strong> raised in isolation units. At 16 months of age, theheifers were artificially inseminated. In six experiments (Fensterbank, 1980) using55 heifers born <strong>to</strong> infected cows, 5 were infected <strong>and</strong> brucellae were isolated duringcalving <strong>and</strong>/or after butchering six weeks later. At 9 <strong>and</strong> 12 months of age, two ofthese animals had serologic titers that were unstable until pregnancy. The other threeheifers did not have serological reactions until the middle or end of pregnancy(latency). The authors of the experiment admit that under natural range conditionsthe frequency of the latency phenomenon could be much lower. In herds in whichvaccination of calves is systematically carried out, the phenomenon may go unnoticed.In a similar vein, other research projects (Lapraik et al., 1975; Wilesmith,1978) have been undertaken on the vertical transmission of brucellosis accompaniedby a prolonged <strong>and</strong> serologically unapparent phase of the infection. In a retrospectivestudy of highly infected herds (Wilesmith, 1978), it was found that 8 of 317heifers (2.5%) born <strong>to</strong> reactive cows tested serologically positive. One study conductedon 150 calves born <strong>to</strong> naturally infected mothers (with positive culture for B.abortus), taken from 82 herds in three southern states in the US, suggests that thelatency phenomenon does not occur very frequently. The calves were raised in isolationuntil sexual maturity <strong>and</strong> breeding. Brucella was not isolated from the progenyof 105 infected cows, nor from the 95 fetuses <strong>and</strong> newborns of these heifers(second generation). Two heifers from the first generation had positive <strong>and</strong> persistentserological reactions from an undetermined source (Ray et al., 1988). The exten<strong>to</strong>f the latency phenomenon is still not known, but it has not prevented the eradicationof bovine brucellosis in vast areas <strong>and</strong> many countries. On the other h<strong>and</strong>, it hasundeniably slowed its eradication in some herds.INFECTION IN SWINE (Figure 6): In swine, the sources of infection are the same asin cattle. The principal routes of transmission are digestive <strong>and</strong> venereal. Contrary<strong>to</strong> the situation in cattle, natural sexual contact is a <strong>common</strong> <strong>and</strong> important mode oftransmission. The infection has often been introduced in<strong>to</strong> a herd following theacquisition of an infected boar. Pigs, because of their eating habits <strong>and</strong> the conditionsin which they are raised, are very likely <strong>to</strong> become infected through the oralroute. It is also probable that they become infected by aerosols entering via the conjunctivaor upper respira<strong>to</strong>ry tract.INFECTION IN GOATS AND SHEEP (Figure 7): Goats <strong>and</strong> sheep are infected with B.melitensis in a manner similar <strong>to</strong> cattle. The role of the buck <strong>and</strong> ram in transmissionof the infection is not well established. Infection of goats in utero is notunusual, <strong>and</strong> kids can also become infected during the suckling period; such infectionmay persist in some animals.In ram epididymitis caused by B. ovis, semen is the main <strong>and</strong> possibly the onlysource of infection. The infection is <strong>common</strong>ly transmitted from one ram <strong>to</strong> another


54 BACTERIOSESby rectal or preputial contact. Transmission may also occur through the ewe whenan infected ram deposits his semen <strong>and</strong> another ram breeds her shortly thereafter.The infection is not very <strong>common</strong> in ewes, <strong>and</strong> when it occurs it is contracted bysexual contact. B. ovis does not persist very long in ewes <strong>and</strong> is generally eliminatedbefore the next lambing period.Figure 6. Swine brucellosis (Brucella suis). Mode of transmission.BoarSexual contactSowMainly by direct <strong>and</strong>indirect contact, rarely byingestion of raw meatFetuses, fetal membranes,vaginal secretionsManContactEnvironmentalcontaminationIngestion, contactBoarFigure 7. Caprine <strong>and</strong> ovine brucellosis (Brucella melitensis). Mode of transmission.Goats<strong>and</strong>sheepFetuses, fetalmembranes,vaginal secretionsEnvironmentalcontamination(pasture, forage,water)IngestionGoats<strong>and</strong>sheepIngestion of raw milk<strong>and</strong> fresh cheeses; contactContact with fetuses,fetal membranesMan


BRUCELLOSIS 55INFECTION IN DOGS: The transmission of B. canis occurs as a result of contact withvaginal secretions, fetuses, <strong>and</strong> fetal membranes. Infected males may transmit theinfection <strong>to</strong> bitches during coitus. The milk of infected bitches is another possiblesource of infection. Human cases recorded in the literature amount <strong>to</strong> several dozen,many resulting from contact with bitches that had recently aborted.Role of Animals in the Epidemiology of the Disease: The role of animals isessential. Cases of human-<strong>to</strong>-human transmission are exceptional. Brucellosis is azoonosis par excellence.Diagnosis: In man, a clinical diagnosis of brucellosis based on symp<strong>to</strong>ms <strong>and</strong> his<strong>to</strong>ryshould always be confirmed in the labora<strong>to</strong>ry. Isolation <strong>and</strong> typing of the causalagent is definitive <strong>and</strong> may also indicate the source of the infection. Blood or marrowfrom the sternum or ileal crest taken while the patient is febrile is cultured in appropriatemedia. Culture material may also be taken from lymph nodes, cerebrospinalfluid, <strong>and</strong> abscesses. It is recommended that the cultures be repeated several times,especially in enzootic areas of B. abortus. Due <strong>to</strong> the widespread use of antibioticsbefore diagnosis in febrile patients, bacteriologic examinations, particularly of blood,often yield negative results, <strong>and</strong> serologic tests become increasingly necessary. Theserum agglutination test, preferably in tubes, is the simplest <strong>and</strong> most widely usedprocedure. A high titer (more than 100 international units, IU) <strong>and</strong> increasing titersin repeated serum samples provide a good basis for diagnosis. Cross-reactions inserum agglutination have been observed in cases of cholera or tularemia (or as aresult of vaccination against these <strong>diseases</strong>) <strong>and</strong> in infections caused by Yersinia enterocolitica0:9, as well as Escherichia coli 0:157 <strong>and</strong> 0:116, Salmonella serotypes ofKauffmann-White group N, <strong>and</strong> Pseudomonas mal<strong>to</strong>phila (Corbel et al., 1984). Theserum agglutination test reveals both M <strong>and</strong> G immunoglobulins. It is generallyaccepted that in an active stage of brucellosis IgG is always present. Thus, when lowserum agglutination titers are found, tests <strong>to</strong> detect the presence of IgG must be performed,such as the 2-mercap<strong>to</strong>ethanol (ME) <strong>and</strong> complement fixation (CF) test (inman, IgGs fix complement but often lack agglutinating power). These tests are of specialinterest in chronic brucellosis, where active infection may continue even thoughagglutination titers return <strong>to</strong> low levels. The intradermal test with noncellular allergensis useful for epidemiological studies, but not for clinical diagnosis.The 2-mercap<strong>to</strong>ethanol test is also useful in following the treatment <strong>and</strong> cure ofthe patient. In one study (Buchanan <strong>and</strong> Faber, 1980), the titers of 92 brucellosispatients were followed for 18 months with tube agglutination <strong>and</strong> ME tests. Despiteantibiotic treatment, the tube agglutination test continued positive for 18 months in44 (48%) of the patients, but the ME titers were positive in only 8 (9%) of thepatients at the end of one year <strong>and</strong> in 4 (4%) at 18 months. None of the 84 patientstesting negative by ME at the end of a year of treatment had signs or symp<strong>to</strong>ms ofbrucellosis <strong>and</strong> none acquired chronic brucellosis. By contrast, four of the eightpatients testing positive by ME after a year continued <strong>to</strong> have symp<strong>to</strong>ms of brucellosis<strong>and</strong> had <strong>to</strong> continue treatment. Thus, a negative result by ME provides goodevidence that a patient does not have chronic brucellosis <strong>and</strong> that the antibiotic treatmentwas successful. If effective treatment is begun early, it is possible that IgG antibodies(resistant <strong>to</strong> ME) never develop. This was probably the case in three patientswho acquired brucellosis in the labora<strong>to</strong>ry <strong>and</strong> in whom infection was confirmed byblood culture. Diagnosis <strong>and</strong> treatment were done early enough that at no time dur-


56 BACTERIOSESing the two-year follow-up did these patients show ME-resistant antibodies (García-Carrillo <strong>and</strong> Col<strong>to</strong>rti, 1979). However, other researchers dispute the usefulness ofthis test in the diagnosis of brucellosis (Díaz <strong>and</strong> Moriyon, 1989).Other useful methods for the diagnosis of human brucellosis are the rose bengaltest <strong>and</strong> counterimmunoelectrophoresis. The rose bengal test is easily performed <strong>and</strong>is recommended over the plate agglutination test or the Huddleson method. In astudy of 222 cases (Díaz et al., 1982), rose bengal was the most sensitive test, with98.3% positive results. Counterimmunoelectrophoresis was positive in 84.9% of theacute cases <strong>and</strong> in 91.6% of the chronic cases.The indirect enzyme-linked immunosorbent assay (ELISA) test has been used forsome years with good results in terms of specificity <strong>and</strong> sensitivity in research (Díaz<strong>and</strong> Moriyón, 1989). It is a very versatile test <strong>and</strong>, once it is introduced in a labora<strong>to</strong>ry,it can be adapted for use with many other <strong>diseases</strong>.The Joint FAO/<strong>WHO</strong> Expert Committee on Brucellosis (<strong>WHO</strong>, 1986) calls attention<strong>to</strong> the limited value of serological tests in individuals who are repeatedly exposed<strong>to</strong> brucellae because they can be serologically positive in the absence of symp<strong>to</strong>ms.This category would include veterinarians, vaccina<strong>to</strong>rs, <strong>and</strong> labora<strong>to</strong>ry personnelinvolved in the production of antigens, vaccines, <strong>and</strong> cultures of clinical specimens.In serologic diagnosis of humans or animals, it is necessary <strong>to</strong> bear in mind thatat the outset of the infection only IgM antibodies are produced; consequently, theagglutination test will provide the best st<strong>and</strong>ard for diagnosis, since ME will yieldnegative results. As the infection progresses, IgG antibodies resistant <strong>to</strong> the ME testwill appear <strong>and</strong> will increase unless appropriate treatment is begun.Diagnosis of infection caused by B. melitensis, B. suis, <strong>and</strong> B. abortus is carriedout with a properly st<strong>and</strong>ardized antigen of B. abortus (Al<strong>to</strong>n et al., 1976). However,this antigen does not permit diagnosis of infection caused by B. canis, since thisspecies of Brucella (as well as B. ovis) is found in a rugose (R) phase, lacking thelipopolysaccharidic surface that characterizes “classic brucellae” (for diagnosis ofB. canis <strong>and</strong> B. ovis, see below).In cattle, the diagnosis is based primarily on serology. A great many serologictests are presently available, all of which are useful when applied with judgment.Both a serologic test reaction <strong>and</strong> the test’s usefulness in each circumstance are aresult of the sensitivity it shows <strong>to</strong> antibodies of different immunoglobulin types <strong>and</strong>of the seric concentration of each type of antibody (Chappel et al., 1978). The mostthoroughly studied immunoglobulins in bovine brucellosis are IgM <strong>and</strong> IgG.Although available tests are not qualitative enough <strong>to</strong> identify an individualimmunoglobulin, they do indicate which one predominates. In the diagnosis ofbovine brucellosis, the evolution of immunoglobulins during infection <strong>and</strong> vaccinationis of special interest. In both cases, the IgMs appear first, followed by the IgGs.The difference is that in infected animals, the IgGs tend <strong>to</strong> increase <strong>and</strong> persist,while in calves vaccinated at between three <strong>and</strong> eight months, the IgGs tend <strong>to</strong> disappearabout six months after vaccination. Based on this fact, complementary testsare used <strong>to</strong> distinguish infection from the agglutination titer, which may persist aftervaccination with strain 19, <strong>and</strong> also from heterospecific reactions caused by bacteriathat share surface antigens with the brucellae <strong>and</strong> that give rise <strong>to</strong> antibodies that,in general, are the IgM type.According <strong>to</strong> their use in different countries, serologic tests may be classified asfollows: (1) routine or operative, (2) complementary, (3) epidemiological surveil-


BRUCELLOSIS 57lance, <strong>and</strong> (4) screening tests. A single test might serve as operative, as diagnosticallydefinitive, as a screen, or as complementary, depending on the program employing it.Serum agglutination tests (tube <strong>and</strong> plate) have been <strong>and</strong> continue <strong>to</strong> be widelyused. They contributed greatly <strong>to</strong> the reduction of infection rates in Europe, Australia,<strong>and</strong> the Americas. Nevertheless, when the proportion of infected herds <strong>and</strong> worldwideprevalence is reduced, their limitations become apparent in so-called “problem”herds <strong>and</strong> it becomes necessary <strong>to</strong> use other tests <strong>to</strong> help eradicate the infection. Thetests are internationally st<strong>and</strong>ardized, easy <strong>to</strong> carry out, <strong>and</strong> allow the examination ofa great many samples. In agglutination tests, the IgM reaction predominates. In animalsclassified as suspect or marginally positive, complementary tests are used <strong>to</strong>clarify their status. However, it is necessary <strong>to</strong> keep in mind that low agglutinationtiters could be due <strong>to</strong> recent infection <strong>and</strong> it is thus advisable <strong>to</strong> repeat the test.The rose bengal test (with buffered antigen) is fast, easy, <strong>and</strong> allows processing ofmany samples per day. It is qualitative <strong>and</strong> classifies animals as positive or negative.In regions where incidence of infection is low or where systematic vaccination ofcalves is practiced, the rose bengal test gives many “false positives,” <strong>and</strong> so is unspecificif used as the only <strong>and</strong> definitive test. In many countries, such as Great Britain<strong>and</strong> Australia, it is used as a preliminary or screening test. Animals showing a negativetest result are so classified <strong>and</strong> those testing positive are subjected <strong>to</strong> other testsfor confirmation. In regions of high incidence, results are very satisfac<strong>to</strong>ry. Rosebengal may also be used as a complementary test for those animals classified as suspectby agglutination. Many suspect sera test negative <strong>to</strong> rose bengal, <strong>and</strong> since thistest is very sensitive (there are few “false negatives”) <strong>and</strong> detects the infection early,there is little risk of missing infected animals.The principal complementary tests are complement fixation, 2-mercap<strong>to</strong>ethanol,<strong>and</strong> rivanol. Other tests have been developed, such as indirect hemolysis, ELISA fordifferent types of immunoglobulins, <strong>and</strong> radial immunodiffusion with a polysaccharideantigen. All these tests are used <strong>to</strong> distinguish antibodies caused by the infectionfrom those left by vaccination or stimulated by heterospecific bacteria.Both the direct <strong>and</strong> the competitive ELISA tests are appropriate for diagnosis ofbrucellosis in all species according <strong>to</strong> the consensus of groups of experts that havemet several times in Geneva. <strong>WHO</strong>, with the collaboration of FAO, the InternationalOrganization of Epizootics, the International A<strong>to</strong>mic Energy Agency, <strong>and</strong> theseorganizations’ reference labora<strong>to</strong>ries, is coordinating a project <strong>to</strong> evaluate <strong>and</strong> st<strong>and</strong>ardizethese assays as well as the antigens <strong>and</strong> other technical variables.In Australia, the ELISA technique <strong>and</strong> the complement fixation test have beenvery useful in recent phases in the eradication of bovine brucellosis, when many“problem herds” occur with “latent carrier animals.” In comparison with the CF test,ELISA revealed a significantly higher number of reactive animals in infected herds,both vaccinated (with strain 19) <strong>and</strong> unvaccinated, but gave negative results in herdsfree of brucellosis, whether vaccinated or not. The specificity of ELISA in the groupof infected herds was less than that of CF, but sensitivity—which is what wasneeded—was greater (Cargill et al., 1985). It costs less <strong>to</strong> eliminate some false positiveanimals in the final phase of eradication than <strong>to</strong> allow the infection <strong>to</strong> reassertitself <strong>and</strong> spread in the herd because one or more infected animals remained(Sutherl<strong>and</strong> et al., 1986). The competitive enzymatic immunoassay also lends itself<strong>to</strong> differentiating the reactions of animals vaccinated with strain 19 <strong>and</strong> animals naturallyinfected, using the O polysaccharide antigen (Nielsen et al., 1989).


58 BACTERIOSESThe complement fixation test is considered the most specific, but it is laborious,complicated, <strong>and</strong> involves many steps <strong>and</strong> variables. Moreover, it is not st<strong>and</strong>ardizedinternationally. Other, simpler tests can take its place, such as 2-mercap<strong>to</strong>ethanol<strong>and</strong> rivanol, which measure the IgG antibodies.Animal health labora<strong>to</strong>ries in the US <strong>and</strong> various labora<strong>to</strong>ries in Latin Americahave successfully used the BAPA (buffered antigen plate agglutination) or BPA(buffered plate antigen) screening test, which is performed on a plate with a bufferedantigen at pH 3.65 (Angus <strong>and</strong> Bar<strong>to</strong>n, 1984). BAPA greatly simplifies the workwhen numerous blood samples must be examined, because it eliminates the negativesamples <strong>and</strong> many of the sera with nonspecific reactions. The test results classify thesamples as negative (which are definitively discarded) <strong>and</strong> presumably positive; thelatter are submitted <strong>to</strong> one or more definitive <strong>and</strong>/or complementary tests, such astube agglutination, complement fixation, or 2-mercap<strong>to</strong>ethanol. This test was alsoevaluated in Canada (Stemshorn et al., 1985) <strong>and</strong> Argentina (González Tomé et al.,1989) with very favorable results.Epidemiological surveillance of brucellosis is carried out separately on dairy <strong>and</strong>meat-producing herds, at strategic checkpoints <strong>and</strong> using different diagnostic tests.The principal objective is <strong>to</strong> identify infected herds <strong>and</strong> moni<strong>to</strong>r healthy ones. Forbeef cattle, screening tests or other tests of presumed high sensitivity are used, suchas BAPA, <strong>and</strong> the checkpoints for collecting samples are cattle markets <strong>and</strong> slaughterhouses.The sera that test positive are then subjected <strong>to</strong> st<strong>and</strong>ard tests <strong>and</strong> theanimals are traced back <strong>to</strong> their points of origin. For dairy cattle, the milk-ring testis used. It is very simple <strong>and</strong> allows the examination of many herds in a short time.The composite samples are gathered from milk cans or tanks at collection points<strong>and</strong> dairy plants or on the dairy farm itself. If a positive sample is found, individualserologic examinations of the animals belonging <strong>to</strong> the source herd must thenbe carried out.Bacteriologic examinations are of more limited use. The samples most oftentested in this way are taken from fetuses, fetal membranes, vaginal secretions, milk,<strong>and</strong> semen. Infected cows may or may not abort, but a high percentage will eliminatebrucellae from the genital tract beginning a few days before parturition <strong>and</strong>continuing some 30 days afterwards. It is estimated that 85% of recently infectedcows <strong>and</strong> more than 15% of chronically infected cows eliminate brucellae duringcalving. Since elimination through milk may be constant or intermittent, milk canbe an excellent material for the isolation of Brucella if examinations are repeated.Serologic testing of bulls should be done using blood serum <strong>and</strong> seminal fluid.Bacteriologic examination of semen should be repeated if results are negative, sincebrucellae may be shed intermittently.In swine, serologic tests are not indicated for individual diagnosis but rather <strong>to</strong>reveal the presence of herd infection. Agglutination (tube or plate), complement fixation,buffered-acid antigen (rose bengal), or BAPA tests may be used. The latter ispreferable because it is negative in herds having only low <strong>and</strong> nonspecific agglutination(tube or plate) titers. For a herd <strong>to</strong> be classified as positive with the agglutinationtest (tube or plate), there must be one or more animals with titers of 100 IUor more.In goats, serologic tests are also applied on a flock basis <strong>and</strong> not on individual animals.In infected flocks, one or more individuals are found with titers of 100 IU ormore; in such cases, titers of 50 IU should be adopted as indicative of infection. The


BRUCELLOSIS 59complement fixation test is considered superior <strong>to</strong> the agglutination test, especiallyin herds vaccinated with B. melitensis Rev. 1, where agglutinating antibodies persistfor long periods. The 2-mercap<strong>to</strong>ethanol test has also given very good results in vaccinatedflocks. The results from the buffered-acid antigen (rose bengal) test arepromising, but experience with it is limited <strong>and</strong> definitive conclusions cannot bedrawn at this time. The ELISA test is the most promising.In diagnosing infections caused by B. melitensis in sheep, the Coombs’ test(antiglobulin test) modified by Hajdu can reveal 70% of infected animals. The othertests (agglutination, complement fixation) give less satisfac<strong>to</strong>ry results. In using theagglutination <strong>and</strong> complement fixation tests, adoption of significant titer levelslower than those for other animal species is recommended. Counterimmunoelectrophoresiswould detect antibodies against intracellular antigens that appear late in theserum but which remain a long time. Consequently, its use would be appropriate forsheep with chronic brucellosis that test negative by agglutination, rose bengal, <strong>and</strong>complement fixation (Trap <strong>and</strong> Gaumont, 1982). Experts agree that the diagnosticmethods for brucellosis caused by B. melitensis in goats <strong>and</strong> sheep leave much <strong>to</strong> bedesired <strong>and</strong> that more attention should be given <strong>to</strong> this problem given its publichealth importance.In diagnosing ram epididymitis caused by B. ovis, antigen prepared with thisagent must be used. The preferred tests are gel diffusion, complement fixation, <strong>and</strong>ELISA. A study conducted in Australia in flocks infected by B. ovis <strong>and</strong> flocks freeof infection showed that this enzyme immunoassay detected more reactive animals<strong>and</strong> that the complement fixation test failed <strong>to</strong> detect some rams that excreted B.ovis. In infection-free flocks, both ELISA <strong>and</strong> CF produced false positives at a rateof 0.5% (Lee et al., 1985). Bacteriologic examination of semen is an appropriatediagnostic method, but it should be kept in mind that the shedding of brucellae canbe intermittent.For dogs infected by B. canis, the surest diagnostic method is isolation of the etiologicagent from blood, vaginal discharges, milk, or semen, or from fetal tissue <strong>and</strong>placenta. Bacteremia can last from one <strong>to</strong> two years, but after the initial phase it maybecome intermittent; thus, a negative blood culture does not exclude the possibilityof brucellosis.The most <strong>common</strong> serologic tests are plate <strong>and</strong> tube agglutination using B. canisantigen, immunodiffusion in agar gel with antigens extracted from the cell wall, 2-mercap<strong>to</strong>ethanol plate agglutination, <strong>and</strong> the modified 2 ME tube agglutination test.Possibly the most specific test <strong>to</strong> date, but also the least sensitive, is the immunodiffusiontest in agar gel that utilizes antigens extracted from the cy<strong>to</strong>plasm of B.canis. To a greater or lesser degree, all these tests give nonspecific reactions. Zoha<strong>and</strong> Carmichael (1982) showed that the immunodiffusion test using sonicated antigens(internal cellular antigens) is satisfac<strong>to</strong>ry shortly after the onset of bacteremia<strong>and</strong> can detect infected animals for up <strong>to</strong> six months after it disappears, i.e., whenother tests give equivocal results. A new test has been developed that uses a nonmucoid(M–) variant of B. canis as the antigen in tube agglutination, after treatingthe sera with 2 ME. The test is more specific without reducing sensitivity(Carmichael <strong>and</strong> Joubert, 1987).Control: The most rational approach for preventing human brucellosis is the control<strong>and</strong> elimination of the infection in animal reservoirs, as has been demonstrated


60 BACTERIOSESin various countries in Europe <strong>and</strong> the Americas. Some human populations may beprotected by m<strong>and</strong>a<strong>to</strong>ry milk pasteurization. In many goat- <strong>and</strong> sheep-herdingregions, pasteurization of milk is an unattainable goal for the time being. Preventionof the infection in occupational groups (cattlemen, abat<strong>to</strong>ir workers, veterinarians,<strong>and</strong> others who come in<strong>to</strong> contact with animals or their carcasses) is more difficult<strong>and</strong> should be based on health education, the use of protective clothing wheneverpossible, <strong>and</strong> medical supervision.Protecting refrigera<strong>to</strong>r plant <strong>and</strong> slaughterhouse workers against brucellosis isparticularly important because they constitute the occupational group at highest risk.Protection is achieved by separating the slaughter area from other sections <strong>and</strong> controllingair circulation. In countries with eradication programs, slaughter of reactiveanimals is limited <strong>to</strong> one or more designated slaughterhouses (cold s<strong>to</strong>rage plants)with official veterinary inspection in each region. These animals are butchered at theend of the workday with special precautions <strong>and</strong> proper supervision <strong>to</strong> protect theworkers. Employees should be instructed in personal hygiene <strong>and</strong> provided with disinfectants<strong>and</strong> protective clothing. A 5% solution of chloramine or an 8% <strong>to</strong> 10%solution of caustic soda should be used <strong>to</strong> disinfect installations after slaughter(Elberg, 1981). Instruments should be sterilized in an au<strong>to</strong>clave or boiled for 30minutes in a 2% solution of caustic soda. Clothes may be disinfected with a 2%solution of chloramine or a 3% solution of carbolic acid soap followed by washing.H<strong>and</strong>s should be soaked for five minutes in a solution of 1% chloramine or 0.5%caustic soda, <strong>and</strong> then washed with soap <strong>and</strong> water.The immunization of high-risk occupational groups is practiced in the formerSoviet Union <strong>and</strong> China. In the former Soviet Union, good results have apparentlybeen obtained with the use of a vaccine prepared from strain 19-BA of B. abortus(derived from strain 19 used for bovine brucellosis), applied by skin scarification.Annual revaccination is carried out for those individuals not reacting <strong>to</strong> serologic orallergenic tests. To avoid the discomfort caused by the vaccine in man, a vaccine wasrecently developed that consists of chemically defined fractions of the lipid-polysaccharide(LPS) component of the strain 19-BA (Drannovskaia, 1991). In China,an attenuated live vaccine made from B. abortus strain 104M is applied percutaneously.These vaccines are not used in other countries because of possible sideeffects. Promising trials have also been conducted in France with antigenic fractionsof Brucella.Vaccination is recommended for control of bovine brucellosis in enzootic areaswith high prevalence rates. The vaccine of choice is B. abortus strain 19, confirmedby its worldwide use, the protection it gives for the useful lifetime of the animal, <strong>and</strong>its low cost. To avoid interference with diagnosis, it is recommended that vaccinationbe limited (by legislation) <strong>to</strong> young animals (calves of 3 <strong>to</strong> 8 months), as theseanimals rapidly lose the antibodies produced in response <strong>to</strong> the vaccine. It is estimatedthat 65% <strong>to</strong> 80% of vaccinated animals remain protected against the infection.The antiabortive effect of the vaccine is pronounced, thus reducing one of theprincipal sources of infection, the fetuses. In a systematic vaccination program, thebest results are obtained with 70% <strong>to</strong> 90% annual coverage in calves of the properage for vaccination. Male calves <strong>and</strong> females over 8 months of age should not bevaccinated. Where possible, the upper limit should be 6 months. Revaccination isnot recommended. The main objective of systematic <strong>and</strong> m<strong>and</strong>a<strong>to</strong>ry vaccination ofcalves in a given area or country is <strong>to</strong> reduce the infection rate <strong>and</strong> obtain herds


BRUCELLOSIS 61resistant <strong>to</strong> brucellosis, so that eradication of the disease may then begin. It is estimatedthat 7 <strong>to</strong> 10 years of systematic vaccination are necessary <strong>to</strong> achieve thisobjective.In regions or countries with a low prevalence of the disease, an eradication programcan be carried out by repeated serologic diagnostic tests applied <strong>to</strong> the entireherd, <strong>and</strong> elimination of reac<strong>to</strong>rs until all foci of infection have disappeared. Thisprocedure can be used alone (in countries with a low prevalence) or in combinationwith the vaccination of calves. Epidemiological surveillance <strong>and</strong> control of animaltransport are very important in such programs. Countries that are close <strong>to</strong> eradicationmay suspend vaccination with strain 19 or any other vaccine.Until a few years ago, the vaccination of adult cows with strain 19 was inadvisablebecause of the prolonged resistance of antibodies that could interfere withdiagnosis. In the 1950s, several researchers proved that vaccination of adult animalswith a smaller dose could impart an immunity comparable <strong>to</strong> that of a full dose,while at the same time agglutination titers stayed lower <strong>and</strong> disappeared faster. In1975, Nicoletti (1976) began a series of studies in the US using a reduced dose inhighly infected herds; he concluded that vaccination decreases the spread of theinfection within the herd, that antibodies disappear approximately six months aftervaccination, <strong>and</strong> that less than 1% of the females remained infected by the vaccinestrain from three <strong>to</strong> six months after vaccination. Complementary tests were veryuseful in distinguishing between reactions due <strong>to</strong> infection <strong>and</strong> those due <strong>to</strong> vaccination.Other studies, done under both controlled <strong>and</strong> natural conditions, confirmedthese findings (Nicoletti et al., 1978; Al<strong>to</strong>n et al., 1980; Viana et al., 1982; Al<strong>to</strong>n etal., 1983). Vaccination of adult females may be considered in herds suffering acutebrucellosis characterized by abortions <strong>and</strong> rapidly spreading infections, as well as inlarge herds where chronic brucellosis has proven hard <strong>to</strong> eradicate. The recommendeddose is one <strong>to</strong> three billion cells of strain 19 Brucella administered subcutaneously.Only animals testing negative should be vaccinated <strong>and</strong> they should beindelibly marked under government supervision. At the beginning of the operation,reac<strong>to</strong>rs should be eliminated immediately. Vaccinated animals should be examinedserologically 6 months later, using tests such as rivanol, mercap<strong>to</strong>ethanol, <strong>and</strong> complementfixation, <strong>and</strong> those that have become infected should be slaughtered. Usingperiodic serologic examination, it is estimated that a problem herd can be free ofinfection in 18 <strong>to</strong> 24 months (Bar<strong>to</strong>n <strong>and</strong> Lomme, 1980).The control of swine brucellosis consists of identifying <strong>and</strong> certifying brucellosisfreeherds. If infection is diagnosed in an establishment where pigs are raised formarket, it is advisable <strong>to</strong> send the entire herd <strong>to</strong> the abat<strong>to</strong>ir <strong>and</strong> reestablish it withanimals from a brucellosis-free herd. If the infected pigs are valuable for breedingor research, suckling pigs should be weaned at 4 weeks <strong>and</strong> raised in facilities separatefrom the main herd. Periodic serologic tests (such as rose bengal) are recommended<strong>to</strong> eliminate any reac<strong>to</strong>r. Finally, when no brucellosis is found in the newherd <strong>and</strong> it is well established, the original herd should be sent <strong>to</strong> slaughter. Thereare no effective vaccines for swine.Control of the infection caused by B. melitensis in goats <strong>and</strong> sheep is basedmainly on vaccination. The preferred vaccine is B. melitensis Rev. 1, which isadministered <strong>to</strong> 3- <strong>to</strong> 6-month-old females. Adult females can receive a smaller dose(20,000 times fewer bacterial cells than in the dose for young females) of the samevaccine (Al<strong>to</strong>n et al., 1972).


62 BACTERIOSESAs goats are generally raised in marginal areas where socioeconomic conditionsare very poor, it is difficult <strong>to</strong> carry out eradication programs. In these areas, reinfectionoccurs constantly, flocks are often nomadic, <strong>and</strong> animal-raising practicesmake sanitary control difficult. Another important fac<strong>to</strong>r is that diagnostic methodsfor small ruminants are deficient. Experience with Rev. 1 vaccine in Italy, Turkey,Iran, Mongolia, Peru, <strong>and</strong> the Caucasian Republics of the former Soviet Union hasproven it <strong>to</strong> be an excellent means of control. In Mongolia, 6 million animals werevaccinated between 1974 <strong>and</strong> 1977; as a result, the prevalence of from 3 <strong>to</strong> 4 per10,000 animals was reduced by half or more, as was the incidence of human cases.In Malta, after seven years of vaccination of small ruminants with Rev. 1, the numberof human cases per year fell from 260 <strong>to</strong> 29. The same thing happened in Italy,although there are no reference data (Al<strong>to</strong>n, 1987). However, the control procedureof diagnosing <strong>and</strong> sacrificing reac<strong>to</strong>r animals has produced satisfac<strong>to</strong>ry results inareas of low prevalence.Rev. 1 vaccine has some limitations, such as residual virulence, the possibility ofabortions when pregnant females are vaccinated, <strong>and</strong> the limited stability of the vaccine,which necessitates constant moni<strong>to</strong>ring. These disadvantages should not eliminateuse of the vaccine as the basis for control of brucellosis in small ruminants, atleast until there is a better vaccine. A Chinese strain of B. suis biovar 1, known as B.suis strain 2, is considered reliable. This strain was isolated from a swine fetus <strong>and</strong>its virulence was attenuated by continuous <strong>and</strong> repeated replications in culturemedia over years, reaching an attenuation that remains stable. Vaccine B. suis strain2 has been used in China with very good results for more than 20 years, not only insmall ruminants but in cattle <strong>and</strong> swine as well. Its use began in the semiarid regionsof northern China, where vaccination operations were very difficult due <strong>to</strong> the lackof fetters <strong>and</strong> traps, <strong>and</strong> thus the vaccine was administered in the drinking water(Xin, 1986). Various research institutes have conducted tests on conjunctive, oral(with syringes of the type used <strong>to</strong> administer antiparasitic agents), <strong>and</strong> subcutaneousvaccines in small ruminants; it has generally been possible <strong>to</strong> confirm the resultsobtained in China. Elimination of the vaccine strain in milk or through the vaginahas not been confirmed <strong>and</strong> studies continue on this vaccine.Ram epididymitis can be successfully controlled by a combination of the followingmeasures: elimination of rams with clinically recognizable lesions, eliminationof clinically normal rams positive <strong>to</strong> the gel diffusion or the complement fixationtest, <strong>and</strong> separation of young rams (those not yet used for breeding) from adultmales. In some countries, such as New Zeal<strong>and</strong> <strong>and</strong> the US, a bacterin prepared fromB. ovis <strong>and</strong> adjuvants is used. Animals are vaccinated when weaned, revaccinatedone or two months later <strong>and</strong> annually thereafter. This vaccine produces antibodiesagainst B. ovis but not B. abortus. The B. melitensis Rev. 1 vaccine is effectiveagainst epididymitis, but also produces B. abortus antibodies, which could be confusedwith infection by B. melitensis. The B. suis strain 2 vaccine does not provideprotection against ram epididymitis.Brucellosis caused by B. canis in dog kennels can be controlled by repeated serologictests <strong>and</strong> blood cultures, followed by elimination of reac<strong>to</strong>r animals. No vaccinesare available yet. Veterinary clinics should advise owners of the risk of keepinga dog with brucellosis <strong>and</strong> should recommend that the dog be put <strong>to</strong> sleep.


BRUCELLOSIS 63BibliographyAcosta, M., H. Ludueña, D. Barre<strong>to</strong>, M. Moro. Brucellosis en alpacas. Rev Invest Pec1:37–49, 1972.Al-Khalaf, S., A. El-Khaladi. Brucellosis in camels in Kuwait. Com Immun MicrobiolInfect Dis 12:1–4, 1989.Al<strong>to</strong>n, G.G. The epidemiology of Brucella melitensis infection in sheep <strong>and</strong> goats. In:Verger, J.M., M. Plommet, eds. Brucella melitensis. Seminar held in Brussels, 14–15November 1984. Dordrecht: Martinus Nijhoff; 1985.Al<strong>to</strong>n, G.G. Control of Brucella melitensis infection in sheep <strong>and</strong> goats—A review. TropAnim Health Prod 19:65–74, 1987.Al<strong>to</strong>n, G.G., L.A. Corner, P.P. Plackett. Vaccination of pregnant cows with low doses ofBrucella abortus strain 19 vaccine. Aust Vet J 56:369–372, 1980.Al<strong>to</strong>n, G.G., L.A. Corner, P.P. Plackett. Vaccination of cattle against brucellosis. Reduceddoses of strain 19 compared with one <strong>and</strong> two doses of 45/20 vaccine. Aust Vet J 60:175–177,1983.Al<strong>to</strong>n, G.G., L.M. Jones, C. García-Carrillo, A. Trenchi. Brucella melitensis Rev. 1 <strong>and</strong>Brucella abortus 45/20 vaccines in goats: Immunity. Am J Vet Res 33:1747–1751, 1972.Al<strong>to</strong>n, G.G., L.M. Jones, D.E. Pietz. Labora<strong>to</strong>ry techniques in brucellosis. 2nd ed. Geneva:World Health Organization; 1975. (Monographs Series 55).Anczykowski, F. Further studies on fowl brucellosis. II. Labora<strong>to</strong>ry experiments. Pol ArchWet 16:271–292, 1973.Angus, R.D., C.E. Bar<strong>to</strong>n. The production <strong>and</strong> evaluation of a buffered plate antigen for usein a presumptive test for brucellosis. In: Third International Symposium on Brucellosis,Algiers, Algeria, 1983. Developments in Biological St<strong>and</strong>ardization. Basel: Karger; 1984.Banai, M., I. Mayer, A. Cohen. Isolation, identification, <strong>and</strong> characterization in Israel ofBrucella melitensis biovar 1 atypical strains susceptible <strong>to</strong> dyes <strong>and</strong> penicillin, indicating theevolution of a new variant. J Clin Microbiol 28:1057–1059, 1990.Barg, L. Isolamen<strong>to</strong> de Brucella canis em Minas Gerais, Brazil. Pesquisa de aglutininas emsoros caninos e humanos [thesis]. Belo Horizonte: Universidad Federal de Minas Gerais, 1975.Bar<strong>to</strong>n, C.E., J.R. Lomme. Reduced-dose whole herd vaccination against brucellosis: Areview of recent experience. J Am Vet Med Assoc 177:1218–1220, 1980.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Buchanan, T.M., L.C. Faber. 2-mercap<strong>to</strong>ethanol brucella agglutination test: Usefulness forpredicting recovery from brucellosis. J Clin Microbiol 11:691–693, 1980.Buchanan, T.M., S.L. Hendricks, C.M. Pat<strong>to</strong>n, R.A. Feldman. Brucellosis in the UnitedStates. An abat<strong>to</strong>ir-associated disease. Part III. Epidemiology <strong>and</strong> evidence for acquiredimmunity. Medicine 53:427–439, 1974.Cargill, C., K. Lee, I. Clarke. Use of an enzyme-linked immunosorbent assay in a bovinebrucellosis eradication program. Aust Vet J 62:49–52, 1985.Carmichael, L.E., J.C. Joubert. A rapid agglutination test for the serodiagnosis ofBrucella canis infection that employs a variant (M–) organism as antigen. Cornell Vet77:3–12, 1987.Chappel, R.J., D.J. McNaught, J.A. Bourke, G.S. Allen. Comparison of the results of someserological tests for bovine brucellosis. J Hyg 80:365–371, 1978.Chile, Ministerio de Agricultura, Servicio Agrícola <strong>and</strong> Ganadero. Brucellosis bovina,ovina <strong>and</strong> caprina. Diagnóstico, control, vacunación. Paris: Office International desEpizooties; 1987. (Technical Series 6).Corbel, M.J. The serological relationship between Brucella spp., Yersinia enterocoliticaserotype IX <strong>and</strong> Salmonella serotypes of Kauffman-White group. J Hyg 75:151–171, 1975.


64 BACTERIOSESCorbel, M.J., F.A. Stuart, R.A. Brewer. Observations on serological cross-reactionsbetween smooth Brucella species <strong>and</strong> organisms of other genera. In: Third InternationalSymposium on Brucellosis, Algiers, Algeria, 1983. Developments in BiologicalSt<strong>and</strong>ardization. Basel: Karger; 1984.Corbel, M.J., E.L. Thomas, C. García-Carrillo. Taxonomic studies on some atypical strainsof Brucella suis. Br Vet J 140:34–43, 1984.De Lord, V., S. Nie<strong>to</strong>, E. S<strong>and</strong>oval et al. Brucellosis en caprinos: estudios serológicos <strong>and</strong>bacteriológicos en Venezuela. Vet Trop 12:27–37, 1987.De la Vega, E., C. García-Carrillo, C. Arce. Infección natural por Brucella en comadrejasDidelphis marsupialis en la República Argentina. Rev Med Vet 60:283–286, 1979.Díaz, R., E. Maravi-Poma, J.L. Fernández, S. García-Merlo, A. Rivero-Puente. Brucellosis:estudio de 222 casos. Parte IV: Diagnóstico de la brucellosis humana. Rev Clin Esp166:107–110, 1982.Díaz, R., I. Moriyon. Labora<strong>to</strong>ry techniques in the diagnosis of human brucellosis. In:Young, E.J., M.J. Corbel, eds. Brucellosis: Clinical <strong>and</strong> Labora<strong>to</strong>ry Aspects. Boca Ra<strong>to</strong>n:CRC Press; 1989.Drannovskaia, E. Brucella <strong>and</strong> brucellosis in man <strong>and</strong> animals. Izmir, Turkey, 1990. TurkishMicrobiol Soc 16:87–100, 1991.Elberg, S.S. The Brucellae. In: Dubos, R.J., J.G. Hirsch, eds. Bacterial <strong>and</strong> MycoticInfections of Man. 4th ed. Philadelphia <strong>and</strong> Montreal: Lippincott; 1965.Elberg, S.S. Immunity <strong>to</strong> Brucella infection. Medicine 52:339–356, 1973.Elberg, S.S. A guide <strong>to</strong> the diagnosis, treatment <strong>and</strong> prevention of human brucellosis.Geneva: World Health Organization; 1981. VPH/81.31 Rev.1. (Unpublished document).Ewalt, D.R., L.B. Forbes. Atypical isolates of Brucella abortus from Canada <strong>and</strong> theUnited States characterized as dye sensitive with M antigen dominant. J Clin Microbiol25:698–701, 1987.Fensterbank, R. Congenital brucellosis in cattle. Geneva: World Health Organization; 1980.<strong>WHO</strong>/BRUC/80.352. (Unpublished document).Flores-Castro, R., F. Suárez, C. Ramírez-Pfeiffer, L.E. Carmichael. Canine brucellosis:Bacteriological <strong>and</strong> serological investigation of naturally infected dogs in Mexico City. J ClinMicrobiol 6:591–597, 1977.Forbes, L.B. Brucella abortus infection in 14 farm dogs. J Am Vet Med Assoc 196:911–916, 1990.Fredickson, L.E., C.E. Bar<strong>to</strong>n. A serologic survey for canine brucellosis in a metropolitanarea. J Am Vet Med Assoc 165:987–989, 1974.García-Carrillo, C. Mé<strong>to</strong>dos para el diagnóstico de la brucellosis. Gac Vet 32:661–667, 1970.García-Carrillo, C. Programa de erradicación de la brucellosis en California. BuenosAires: Centro Panamericano de Zoonosis, 1975. (Scientific <strong>and</strong> Technical Monographs 9).García-Carrillo, C., E.A. Col<strong>to</strong>rti. Ausencia de anticuerpos resistentes al 2-marcap<strong>to</strong>etanolen tres pacientes de brucellosis. Medicina 39:611–613, 1979.García-Carrillo, C., N.E. Lucero. Brucellosis bovina. Buenos Aires: Hemisferio Sur; 1993.García-Carrillo, C., B. Szyfres, J. González Tomé. Tipificación de brucelas aisladas delhombre <strong>and</strong> los animales en Latin America. Rev Latinoam Microbiol 14:117–125, 1972.George, L.W., L.E. Carmichael. A plate agglutination test for the rapid diagnosis of caninebrucellosis. Am J Vet Res 35:905–909, 1974.Gilman, H.L. Brucellosis. In: Gibbons, W.J., ed. Diseases of Cattle, a text <strong>and</strong> referencework. The work of 54 authors. Whea<strong>to</strong>n: American Veterinary Publication; 1963.González Tomé, J.S., L.J. Villa, E. del Palacio, R. Gregoret. El test de Angus <strong>and</strong> Bar<strong>to</strong>n (BPA)como prueba tamiz en el diagnóstico de la brucellosis bovina. Rev Med Vet 70:34–36, 1989.Hendricks, S.L., M.E. Meyer. Brucellosis. In: Hubbert, W.T., W.F. McCulloch, P.R.Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield:Thomas; 1975.


BRUCELLOSIS 65Kasyanov, A.N., R.G. Aslanyan. Epizootiology <strong>and</strong> clinical appearance of animal brucellosis.In: Lisenko, A., ed. Zoonoses Control. Moscow: VII Centre Projects; 1982.Kaufmann, A.F., M.D. Fox, J.M. Boyce, D.C. Anderson, M.E. Potter, W.J. Mar<strong>to</strong>ne, et al.Airborne spread of brucellosis. Ann NY Acad Sci 353:105–114, 1980.Lapraik, R.D., D.D. Brown, H. Mann. Brucellosis. A study of five calves from reac<strong>to</strong>rdams. Vet Rec 97:52–54, 1975.Lee, K., C. Cargill, H. Atkinson. Evaluation of an enzyme-linked immunosorbent assay forthe diagnosis of Brucella ovis infection in rams. Aust Vet J 62:91–93, 1985.Lu, S-L., J-L. Zhang. Brucellosis in China. In:Young, E.J., M.J. Corbel, eds. Brucellosis:Clinical <strong>and</strong> Labora<strong>to</strong>ry Aspects. Boca Ra<strong>to</strong>n: CRC Press; 1989.Lubani, M., D. Sharda, I. Helin. Probable transmission of brucellosis from breast milk <strong>to</strong> anewborn. Trop Geogr Med 40:151–152, 1988.Luchsinger, D.W., R.K. Anderson. Longitudinal studies of naturally acquired Brucellaabortus infection in sheep. Am J Vet Med Res 40:1307–1312, 1979.Manthei, C.A. Brucellosis as a cause of abortion <strong>to</strong>day. In: Faulkner, L.C., ed. AbortionDiseases of Lives<strong>to</strong>ck. Springfield: Thomas; 1968.Manthei, C.A. Brucellosis. In: Dunne, H.W., ed. Diseases of Swine. 3rd ed. Ames: IowaState University Press; 1970.McCaughey, W.J. Brucellosis in wildlife. In: Diarmid, A., ed. Diseases in Free-living WildAnimals. New York: Academic Press; 1969.McCullough, N.B. Microbial <strong>and</strong> host fac<strong>to</strong>rs in the pathogenesis of brucellosis. In: Mudd,S., ed. Infectious Agents <strong>and</strong> Host Reactions. Philadelphia: Saunders; 1970.Meyer, M.E. Inter- <strong>and</strong> intra-strain variants in the Genus Brucella. In: Third InternationalSymposium on Brucellosis, Algiers, Algeria, 1983. Developments in BiologicalSt<strong>and</strong>ardization. Basel: Karger; 1984.Morgan, W.J.B. Brucella classification <strong>and</strong> regional distribution. In: Third InternationalSymposium on Brucellosis, Algiers, Algeria, 1983. Developments in BiologicalSt<strong>and</strong>ardization. Basel: Karger; 1984.Myers, D.M., L.M. Jones, V.M. Varela-Díaz. Studies of antigens for complement fixation<strong>and</strong> gel diffusion tests in the diagnosis of infections caused by Brucella ovis <strong>and</strong> otherBrucella. Appl Microbiol 23:894–902, 1972.Nicoletti, P. A preliminary report on the efficacy of adult cattle vaccination using strain 19in selected dairy herds in Florida. Proc Annu Meet US Livest Sanit Assoc 80:91–106, 1976.Nicoletti, P., L.M. Jones, D.T. Berman. Adult vaccination with st<strong>and</strong>ard <strong>and</strong> reduced dosesof Brucella abortus strain 19 in a dairy herd infected with brucellosis. J Am Vet Med Assoc173:1445–1449, 1978.Nicoletti, P., F.W. Milward. Protection by oral administration of Brucella abortus strain 19against an oral challenge exposure with a pathogenic strain of Brucella. Am J Vet Res44:1641–1643, 1983.Nielsen, K., J.W. Cherwonogrodzky, J.R. Duncan, D.R. Bundle. Enzyme-linkedimmunosorbent assay for differentiation of the antibody response of cattle naturally infectedwith Brucella abortus or vaccinated with strain 19. Am J Vet Res 50:5–9, 1989.Ossola, A.L., B. Szyfres. Natural infection of sheep by Brucella melitensis in Argentina. AmJ Vet Res 24:446–449, 1963.Pacheco, G., M.T. De Mello. Brucelose. Rio de Janeiro: Institu<strong>to</strong> Brasileiro de Geografia eEstatística; 1956.Pan American Health Organization. Guía para la preparación <strong>and</strong> evaluación de proyec<strong>to</strong>sde lucha contra la brucellosis bovina. Buenos Aires: Centro Panamericano de Zoonosis; 1972.(Technical Note 14).Peres, J.N., A.M. Godoy, L. Barg, J.O. Costa. Isolamen<strong>to</strong> de Brucella canis de carrapa<strong>to</strong>s(Rhipicephalus sanguineus). Arq Esc Vet UFMG 33:51–55, 1981.


66 BACTERIOSESPfischner, W.C.E., K.G. Ishak, E. Neptune, et al. Brucellosis in Egypt: A review of experiencewith 228 patients. Am J Med 22:915, 1957. Cited in: Young, E.J. Clinical manifestationsof human brucellosis. In:Young, E.J., M.J. Corbel, eds. Brucellosis: Clinical <strong>and</strong> Labora<strong>to</strong>ryAspects. Boca Ra<strong>to</strong>n: CRC Press; 1989.Plommet, M., R. Fensterbank. Vaccination against bovine brucellosis with a low dose ofstrain 19 administered by the conjunctival route. III.—Seriological response <strong>and</strong> immunity inthe pregnant cow. Ann Rech Vét 7:9–23, 1976.Plommet, M., R. Fensterbank, G. Renoux, J. Gestin, A. Philippon. Brucellose bovineexpérimentale. XII. Persistance a l’age adulte de l’infection congénitale de la génisse. AnnRech Vét 4:419–435, 1973.Ramacciotti, F. Brucelosis. Córdoba, Argentina: Edición del Au<strong>to</strong>r; 1971.Ray, W.C., R.B. Brown, D.A. Stringfellow, et al. Bovine brucellosis: An investigation oflatency in progeny of culture-positive cows. J Am Vet Med Assoc 192:182–185, 1988.Robertson, F.J., J. Milne, C.L. Silver, H. Clark. Abortion associated with Brucella abortus(biovar 1) in the T.B. mare. Vet Rec 92:480–481, 1973.Ruben, B., J.D. B<strong>and</strong>, P. Wong, J. Colville. Person-<strong>to</strong>-person transmission of Brucellamelitensis. Lancet 337:14–15, 1991.Schwabe, C.W. Veterinary Medicine <strong>and</strong> Human Health. 2nd ed. Baltimore: Williams &Wilkins; 1969.Spink, W.W. The Nature of Brucellosis. Minneapolis: University of Minnesota Press; 1956.Spink, W.W. Brucellosis (Undulant fever, Malta fever). In: Wyngaarden, I.B., L.H. Smith,Jr., eds. Cecil Textbook of Medicine. 16th ed. Philadelphia: Saunders; 1982.Stemshorn, B.W., L.B. Forbes, M.D. Eaglesome, et al. A comparison of st<strong>and</strong>ard serologicaltests for the diagnosis of bovine brucellosis in Canada. Can J Comp Med 49:391–394, 1985.Sutherl<strong>and</strong>, S.S., R.J. Evans, J. Bathgate. Application of an enzyme-linked immunosorbentassay in the final stages of a bovine brucellosis eradication program. Aust Vet J63:412–415, 1986.Szyfres, B. La situación de la brucellosis en Latin America. Bol Hig Epidemiol5:400–409, 1967.Szyfres, B. Taxonomía del género Brucella. Gac Vet 33:28–40, 1971.Szyfres, B., J. González Tomé. Natural Brucella infection in Argentine wild foxes. BullWorld Health Organ 34:919–923, 1966.Szyfres, B., J. González Tomé, T. Palacio Mendieta. Aislamien<strong>to</strong> de Brucella suis dela liebre europea (Lepus europaeus) en la Argentina. Bol Oficina Sanit Panam 65:441–445, 1968.Timm, B.M. Brucellosis. Distribution in Man, Domestic <strong>and</strong> Wild Animals. Berlin:Springer; 1982.Trap, D., R. Gaumont. Comparaison entre electrosynerèse et épreuves serologiques classiquesdans le diagnostic de la brucellose ovine. Ann Rech Vét 13:33–39, 1982.United States, Department of Health <strong>and</strong> Human Services, Centers for Disease Control <strong>and</strong>Prevention (CDC). Annual summary 1981: Reported morbidity <strong>and</strong> mortality in the UnitedStates. MMWR Morb Mort Wkly Rep 30:14, 1982.Van der Hoeden, J. Brucellosis. In: Van der Hoeden, J., ed. Zoonoses. Amsterdam:Elsevier; 1964.Verger, J.M., F. Grimont, P.A.D. Grimont, M. Grayon. Brucella: a monospecific genus asshown by deoxyribonucleic acid hybridization. Int J Syst Bacteriol 35:292–295, 1985.Viana, F.C., J.A. Silva, E.C. Moreira, L.G. Villela, J.G. Mendez, T.O. Dias. Vacinação contrabrucelose bovina com dose reduzida (amostra B 19) por via conjuntival. Arq Esc Vet UFMG34:279–287, 1982.Wilesmith, J.W. The persistence of Brucella abortus infection in calves: A retrospectivestudy of heavily infected herds. Vet Rec 103:149–153, 1978.


CAMPYLOBACTERIOSIS 67Witter, J.F., D.C. O’Meara. Brucellosis. In: Davis, J.W., L.H. Karstady, D.O. Trainer.Infectious Diseases of Wild Mammals. Ames: Iowa State University Press; 1970.World Health Organization (<strong>WHO</strong>). Fifth Report on the World Health Situation,1969–1972. Geneva: <strong>WHO</strong>; 1975. (Official Records 225).World Health Organization (<strong>WHO</strong>). Joint FAO/<strong>WHO</strong> Expert Committee on Brucellosis,Sixth Report. Geneva: <strong>WHO</strong>; 1986. (Technical Report Series 740).Xin, X. Orally administrable brucellosis vaccine: Brucella suis strain 2 vaccine. Vaccine4:212–216, 1986.Young, E.J. Clinical manifestations of human brucellosis. In:Young, E.J., M.J. Corbel, eds.Brucellosis: Clinical <strong>and</strong> Labora<strong>to</strong>ry Aspects. Boca Ra<strong>to</strong>n: CRC Press; 1989.Zoha, S.J., L.E. Carmichael. Serological response of dogs <strong>to</strong> cell wall <strong>and</strong> internal antigensof Brucella canis (B. canis). Vet Microbiol 7:35–50, 1982.


CAMPYLOBACTERIOSISICD-10 A04.5 campylobacter enteritisThe genus Campylobacter (here<strong>to</strong>fore Vibrio) contains several species of importancefor both public <strong>and</strong> animal health. The principal pathogenic species are C.jejuni <strong>and</strong> C. fetus subsp. fetus (previously subsp. intestinalis) <strong>and</strong> C. fetus subsp.venerealis. Occasionally C. coli, C. laridis, <strong>and</strong> C. upsaliensis cause enteritis in man<strong>and</strong> animals. These bacteria are gram-negative, microaerophilic, thermophilic, catalasepositive (with the exception of C. upsaliensis), <strong>and</strong> have a curved or spiral shape.The importance of campylobacteriosis as a diarrheal disease became evidentwhen better knowledge was gained about its requirements for culture <strong>and</strong> isolation,particularly oxygen pressure (strictly microaerophilic) <strong>and</strong> an optimum temperatureof 42°C (thermophilic).Increased medical interest since 1977 in enteritis caused by C. jejuni <strong>and</strong> theenormous bibliography on this new zoonosis make it advisable <strong>to</strong> discuss this <strong>diseases</strong>eparately from those caused by C. fetus <strong>and</strong> its two subspecies. Furthermore,the disease caused by C. jejuni <strong>and</strong> those caused by C. fetus are clinically different.Synonym: Vibrionic enteritis.1. Enteritis caused by Campylobacter jejuniEtiology: Campylobacter jejuni <strong>and</strong> occasionally C. coli. Two principal schemeshave been proposed for serotyping C. jejuni. The scheme proposed by Penner usessomatic antigens <strong>and</strong> includes 60 serotypes, which are identified using the passivehemagglutination method (Penner <strong>and</strong> Hennessy, 1980; McMyne et al., 1982). Thescheme proposed by Lior uses a flaggelar antigen <strong>and</strong> identifies 90 serotypes withthe slide plate agglutination method (Lior, 1982). Pat<strong>to</strong>n et al. (1985) compare bothschemes in 1,405 isolates of human, animal, <strong>and</strong> environmental origin, <strong>and</strong> find that


68 BACTERIOSES96.1% could be typed using the Penner system <strong>and</strong> 92.1% could be typed using theLior system. They also conclude that these schemes complement each other <strong>and</strong> areuseful for epidemiological research.Geographic Distribution: Worldwide.Occurrence in Man: At present, C. jejuni is considered <strong>to</strong> be one of the principalbacterial agents causing enteritis <strong>and</strong> diarrhea in man, particularly in developedcountries. In these countries, the incidence is similar <strong>to</strong> that of enteritis caused bySalmonella. As culture media <strong>and</strong> isolation methods have been perfected, the numberof recorded cases caused by C. jejuni has increased. In Great Britain, the 200public health <strong>and</strong> hospital labora<strong>to</strong>ries had been reporting isolations of salmonellasexceeding those of Campylobacter, but beginning in 1981, the proportions werereversed: 12,496 isolations of Campylobacter as opposed <strong>to</strong> 10,745 of Salmonella(Skirrow, 1982). According <strong>to</strong> Benenson (1990), campylobacteriosis causes 5% <strong>to</strong>14% of diarrhea cases worldwide. Based on records from private medical practice,it has been estimated that 20% of office consultations for enteritis in Great Britainwere associated with campylobacteriosis <strong>and</strong> that there are a projected 600,000cases annually at the national level (Skirrow, 1982). It is harder <strong>to</strong> establish the incidencein developing countries; because of deficiencies in hygiene, C. jejuni is isolatedfrom 5% <strong>to</strong> 17% of persons without diarrhea (Prescott <strong>and</strong> Munroe, 1982) <strong>and</strong>from 8% <strong>to</strong> 31% of persons with diarrhea. Thus, it is likely that Campylobacter isan important cause of infantile diarrhea in the Third World (Skirrow, 1982).The illness affects all age groups. In developing countries, it particularly affectschildren under the age of 2 years; in developed countries, children <strong>and</strong> young adultsbecome ill more frequently. Campylobacteriosis is also an important cause of “travellers’diarrhea” (Benenson, 1990). The disease is primarily sporadic, although thereare also epidemic outbreaks. The largest known epidemics originated from <strong>common</strong>sources, such as unpasteurized milk or contaminated water from the municipal supplyof two European cities. In countries with a temperate climate, the disease is mostprevalent in the warm months.In Great Britain, the US, Canada, <strong>and</strong> Switzerl<strong>and</strong>, consumption of unpasteurizedmilk or products prepared with raw milk has caused campylobacteriosis outbreaks.The largest outbreak in Great Britain affected approximately 3,500 people (Jones etal., 1981). Outbreaks may be due <strong>to</strong> milk contaminated by fecal matter or, less frequently,<strong>to</strong> milk from udders with mastitis caused by C. jejuni. Another outbreakaffected more than 30 people in a small <strong>to</strong>wn in Great Britain. The ensuing investigationshowed that the source of infection was two cows with mastitis caused by C.jejuni that contaminated the bulk milk of 40 cows (Hutchinson et al., 1985). Thesame serotypes of C. jejuni were isolated from the cows, patients’ feces, milk filters,<strong>and</strong> bulk milk.It is estimated that C. jejuni causes more than 90% of human cases of the disease(Karmali <strong>and</strong> Skirrow, 1984) <strong>and</strong> only 10% in other species.Occurrence in Animals: Domestic <strong>and</strong> wild mammals <strong>and</strong> birds constitute thelarge reservoir of C. jejuni,but it is difficult <strong>to</strong> implicate this agent as a cause of diarrhealdisease because a high rate of infection is found in clinically healthy animals.The Disease in Man: Enteritis caused by C. jejuni is an acute illness. In general,the incubation period is from two <strong>to</strong> five days. The principal symp<strong>to</strong>ms are diarrhea,


CAMPYLOBACTERIOSIS 69fever, abdominal pain, vomiting (in one-third of the patients), <strong>and</strong> visible or occultblood (50% <strong>to</strong> 90% of patients). Fever is often accompanied by general malaise,headache, <strong>and</strong> muscle <strong>and</strong> joint pain. The feces are liquid <strong>and</strong> frequently containmucus <strong>and</strong> blood. The course of the illness is usually benign, <strong>and</strong> the patient recoversspontaneously in a week <strong>to</strong> 10 days; acute symp<strong>to</strong>ms often fade in two <strong>to</strong> threedays. Symp<strong>to</strong>ms may be more severe in some patients, similar <strong>to</strong> those of ulcerativecolitis <strong>and</strong> salmonellosis, <strong>and</strong> may lead <strong>to</strong> suspicion of appendicitis <strong>and</strong> anexplora<strong>to</strong>ry lapara<strong>to</strong>my. In some cases, septicemia has been confirmed, either simultaneous<strong>to</strong> the diarrheic illness or afterward. Complications are rare <strong>and</strong> consist ofmeningitis <strong>and</strong> abortions.Enteric campylobacteriosis is a self-limiting disease <strong>and</strong> does not usually requiremedication, except for electrolyte replacement. In cases that require medication,erythromycin is the antibiotic of choice.The Disease in Animals: C. jejuni has been identified as an etiologic agent inseveral illnesses of domestic animals (Prescott <strong>and</strong> Munroe, 1982).CATTLE: Enteritis caused by C. jejuni in calves is clinically similar <strong>to</strong> that in man.Calves suffer a moderate fever <strong>and</strong> diarrhea that may last as long as 14 days. It isalso possible that this agent causes mastitis in cows, as demonstrated by the fact thatexperimental inoculation of the udder with a small number of bacteria causes acutemastitis (see outbreaks due <strong>to</strong> raw milk in the section on occurrence in man).SHEEP: C. jejuni is a major cause of abortions in sheep. In the number of outbreaks,it is assigned a role similar <strong>to</strong> that of C. fetus subsp. fetus (intestinalis).Sheep abort <strong>to</strong>ward the end of their pregnancy or give birth at term <strong>to</strong> either dead orweak lambs that may die within a few days.DOGS AND CATS: Puppies with diarrhea constitute a source of infection for theirowners. Diarrhea is the predominant symp<strong>to</strong>m <strong>and</strong> vomiting seems <strong>to</strong> be frequent.The disease is more frequent in puppies, but may occur in adult animals as well. Foxet al. (1984) described an outbreak caused by C. jejuni in nine of 10 young beaglesthat had diarrheal feces with traces of bile <strong>and</strong> occasionally blood. In Engl<strong>and</strong>, astudy of dogs treated at various veterinary clinics isolated C. jejuni from 59 (11.6%)of the 505 dogs with diarrhea <strong>and</strong> from only 2 (1.6%) of the 122 dogs without diarrhea.In another study (Fleming, 1983), 39 dogs had either persistent or intermittentchronic diarrhea.Burnens <strong>and</strong> Nicolet (1992) cultured 241 samples of fecal matter from dogs <strong>and</strong>156 from cats with diarrhea. The cultures were positive for Campylobacter spp. in20% of the dog samples <strong>and</strong> in 13% of the cat samples. The frequency of C.upsaliensis among the positive cultures was approximately equal in dogs <strong>and</strong> cats.However, the authors present no conclusions regarding the pathogenic role of C.upsaliensis in dogs <strong>and</strong> cats because they did not examine animals without diarrhea.OTHER MAMMALS: Enteritis caused by C. jejuni probably occurs in many other animalspecies. It has been described in monkeys <strong>and</strong> in one outbreak in young horses.FOWL: Fowl are an important reservoir of C. jejuni. Although it was possible <strong>to</strong>cause diarrhea in 3-day-old chicks with orally administered C. jejuni, it is not knownif the illness occurs naturally, since a high proportion of healthy birds harbor thebacteria in their intestines.


70 BACTERIOSESFigure 8. Campylobacteriosis (Campylobacter jejuni). Mode of transmission.Cattle, sheep,dogs, cats,wild <strong>and</strong>domesticmammals<strong>and</strong> birdsFecal routeEnvironmentalcontaminationOral routeCattle, sheep,dogs, cats,wild <strong>and</strong>domesticmammals<strong>and</strong> birdsManSource of Infection <strong>and</strong> Mode of Transmission (Figure 8): Mammals <strong>and</strong> birds,both domestic <strong>and</strong> wild, are the principal reservoir of C. jejuni. In studies by variousauthors (Skirrow, 1982; Prescott <strong>and</strong> Munroe, 1982), C. jejuni was found in thececa of 100% of 600 turkeys <strong>and</strong> in the droppings of 38 out of 46 chickens <strong>and</strong> 83out of 94 ducks that had large numbers of the bacteria in their intestines prior <strong>to</strong>slaughter. The organism has been found in several species of wild birds, for example,in 35% of migra<strong>to</strong>ry birds, 50% of urban pigeons, <strong>and</strong> 20% <strong>to</strong> 70% of seagulls.The agent has been isolated from the feces of 2.5% <strong>to</strong> 100% of healthy cows, fromthe gallbladder of 20 out of 186 sheep, from the feces of 0% <strong>to</strong> 30% of healthy dogs,<strong>and</strong> also from a wide variety of wild mammal species.C. jejuni is <strong>common</strong>ly found in natural water sources, where it can survive forseveral weeks at low temperatures. However, it is interesting <strong>to</strong> note that it hasalways been found in the presence of fecal coliforms, <strong>and</strong> therefore the contaminationpresumably stems from animals (mammals <strong>and</strong> fowl) <strong>and</strong>, in some circumstances,from man. The source of infection is almost always food, although it issometimes difficult <strong>to</strong> identify the immediate source. Given the <strong>common</strong> occurrenceof C. jejuni <strong>and</strong> C. coli in the intestines of mammals <strong>and</strong> fowl (C. jejuni can survivefor several weeks at 4°C on the moist surface of chickens), it can easily be assumedthat contamination of bird <strong>and</strong> animal meat is a frequent occurrence. One study conductedby various labora<strong>to</strong>ries in the US demonstrated that approximately 30% ofthe 300 chickens included in the sample had C. jejuni <strong>and</strong> that 5.1% of the 1,800samples of red meat were contaminated. C. coli was isolated from pork <strong>and</strong> C. jejuniwas isolated from other meats (Stern et al., 1985).


CAMPYLOBACTERIOSIS 71The infection in man may be caused by cross contamination in the kitchen ofmeats with C. jejuni <strong>and</strong> other foods that do not require cooking, or that are undercooked(Griffith <strong>and</strong> Park, 1990). Other sources of infection are unpasteurized milk<strong>and</strong> milk products, river water, <strong>and</strong> inadequately treated municipal water. In somecases, the infection is acquired directly from animals, especially from puppies <strong>and</strong>cats with diarrhea. The victims are almost always children who play with these animals<strong>and</strong> come in contact with the animal’s feces.In peripheral urban areas of Lima (Peru), Grados et al. (1988) studied 104 childrenunder the age of 3 years who had diarrhea <strong>and</strong> compared them <strong>to</strong> the samenumber of children without gastrointestinal disorders (control group) in order <strong>to</strong>identify the various risk fac<strong>to</strong>rs. The authors concluded that the presence of chickens<strong>and</strong> hens in the home environment constitutes an important risk fac<strong>to</strong>r. Childrenbecome infected by contact with the droppings of birds in the household environment.Another interesting fact is that slaughterhouse workers—particularly thosewho come in<strong>to</strong> direct contact with animals <strong>and</strong> their by-products—have a muchhigher rate of positive reactions <strong>to</strong> Campylobacter spp. than the blood donors whoserved as controls (Mancinelli et al., 1987).Person-<strong>to</strong>-person transmission may occur, but is unusual. Among the few casesdescribed was a nosocomial infection of children in Mexico (Flores-Salorio et al.,1983). Untreated patients may eliminate C. jejuni for six weeks, <strong>and</strong> a few for a yearor more. As in the case of other enteric infections, entry is through the digestive tract.Diagnosis: Consists mainly of isolating the agent from the patient’s feces.Diagnosis is made using selective media that are incubated in an atmosphere of 5%oxygen, 10% carbon dioxide, <strong>and</strong> 85% nitrogen, preferably at a temperature of43°C. Serologic diagnosis may be done using direct immunofluorescence or othertests on paired sera.In animals, because of the high rate of healthy carriers, isolation of the agent isinadequate proof that it is responsible for the illness, <strong>and</strong> it is advisable <strong>to</strong> confirman increase in titers with serologic testing.Control: According <strong>to</strong> present knowledge of the epidemiology of the illness, preventivemeasures can be only partial in scope. In a study of risk fac<strong>to</strong>rs in Colorado(USA), where sporadic cases of infection were caused by C. jejuni, it was estimatedthat approximately one-third of the cases could have been prevented by such measuresas avoiding the consumption of untreated water, unpasteurized milk, or undercookedchicken (Hopkins et al., 1984). People in contact with dogs <strong>and</strong> cats withdiarrhea should follow personal hygiene rules, such as thorough h<strong>and</strong>washing. Sickanimals should not be in contact with children. The same recommendations on personalhygiene apply <strong>to</strong> homemakers. In the kitchen, care should be taken <strong>to</strong> separateraw animal products from other foods, particularly in the case of fowl. Control ofthe infection in animals is clearly desirable, but is not presently feasible, given thewide diffusion of the agent <strong>and</strong> its presence in wild animal reservoirs.BibliographyBenenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.


72 BACTERIOSESBurnens, A.P., J. Nicolet. Detection of Campylobacter upsaliensis in diarrheic dogs <strong>and</strong>cats, using a selective medium with cefoperazone. Am J Vet Res 53:48–51, 1992.Fleming, M.P. Association of Campylobacter jejuni with enteritis in dogs <strong>and</strong> cats. Vet Rec113:372–374, 1983.Flores-Salorio, S.G., V. Vázquez-Alvarado, L. Moreno-Altamirano. Campylobacter comoagente etiológico de diarrea en niños. Bol Med Hosp Infant Mex 40:315–319, 1983.Fox, J.G., K.O. Maxwell, J.I. Ackerman. Campylobacter jejuni associated diarrhea in commerciallyreared beagles. Lab Animal Sci 34:151–155, 1984.Grados, O., N. Bravo, R.E. Black, J.P. Butzler. Paediatric campylobacter diarrhoeafrom household exposure <strong>to</strong> live chickens in Lima, Peru. Bull World Health Organ66:369–374, 1988.Griffiths, P.L., R.W. Park. Campylobacters associated with human diarrhoeal disease. Areview. J Appl Bacteriol 69:281–301, 1990.Hopkins, R.S., R. Olmsted, G.R. Istre. Endemic Campylobacter jejuni infection inColorado: Identified risk fac<strong>to</strong>rs. Am J Public Health 74:249–250, 1984.Hutchinson, D.N., F.J. Bol<strong>to</strong>n, P.M. Hinchliffe, et al. Evidence of udder excretion ofCampylobacter jejuni as the cause of milk-borne campylobacter outbreak. J Hyg 94:205–215, 1985.Jones, P.H., A.T. Willis, D.A. Robinson, et al. Campylobacter enteritis associated with theconsumption of free school milk. J Hyg 87:155–162, 1981.Karmali, M.A., M.B. Skirrow. Taxonomy of the genus Campylobacter. In: Butzler, J.P., ed.Campylobacter Infection in Man <strong>and</strong> Animals. Boca Ra<strong>to</strong>n: CRC Press; 1984.Lior, H., D.L. Woodward, J.A. Edgar, et al. Serotyping of Campylobacter jejuni by slideagglutination based on heat-labile antigenic fac<strong>to</strong>rs. J Clin Microbiol 15:761–768, 1982.Mancinelli, S., L. Palombi, F. Riccardi, M.C. Marazzi. Serological study of Campylobacterjejuni infection in slaughterhouse workers [letter]. J Infect Dis 156:856, 1987.McMyne, P.M.S., J.L. Penner, R.G. Mathias, W.A. Black, J.N. Hennessy. Serotyping ofCampylobacter jejuni isolated from sporadic cases <strong>and</strong> outbreaks in British Columbia. J ClinMicrobiol 16:281–285, 1982.Pat<strong>to</strong>n, C.M., T.J. Barrett, G.K. Morris. Comparison of the Penner <strong>and</strong> Lior methods forserotyping Campylobacter spp. J Clin Microbiol 22:558–565, 1985.Penner, J.L., J.N. Hennessy. Passive hemagglutination technique for serotypingCampylobacter fetus subsp. jejuni on the basis of soluble heat-stable antigens. J ClinMicrobiol 12:732–737, 1980.Prescott, J.M., D.L. Munroe. Campylobacter jejuni. Enteritis in man <strong>and</strong> domestic animals.J Am Vet Med Assoc 181:1524–1530, 1982.S<strong>and</strong>stedt, K., J. Ursing. Description of Campylobacter upsaliensis sp.nov. previouslyknown as CNW group. System Appl Microbiol 14:39–45, 1989.Skirrow, M.B. Campylobacter enteritis. The first five years. J Hyg 89:175–184, 1982.Stern, N.J., M.P. Hernández, L. Blakenship, et al. Prevalence <strong>and</strong> distribution of Campylobacterjejuni <strong>and</strong> Campylobacter coli in retail meats. J Food Protect 48:595–599, 1985.2. Diseases caused by Campylobacter fetusSynonyms: Vibriosis, vibrionic abortion, epizootic infertility, bovine genital vibriosis,epizootic ovine abortion.Etiology: Campylobacter (Vibrio) fetus subsp. fetus (intestinalis) <strong>and</strong> C. fetussubsp. venerealis. C. fetus develops in such media as blood agar <strong>and</strong> Brucella agar;it is microaerophilic, but is unlike C. jejuni in that it grows at 25°C but not at 42°C.Geographic Distribution: Worldwide.


CAMPYLOBACTERIOSIS 73Occurrence in Man: Un<strong>common</strong>. Up <strong>to</strong> 1981, the literature recorded at least 134confirmed cases (Bokkenheuser <strong>and</strong> Sutter, 1981), most of them occurring in the US<strong>and</strong> the rest in various parts of the world. The incidence is believed <strong>to</strong> be muchhigher than that recorded.Occurrence in Animals: The disease is <strong>common</strong> in cattle <strong>and</strong> sheep <strong>and</strong> occursworldwide.The Disease in Man: The strains isolated from man have characteristics similar <strong>to</strong>those of C. fetus subsp. fetus (intestinalis), which causes outbreaks of abortion amongsheep <strong>and</strong> sporadic cases in cattle. Two cases caused by C. fetus subsp. venerealis havealso been described (Veron <strong>and</strong> Chatelain, 1973). Campylobacteriosis is generally recognizedwhen accompanied by predisposing debilitating fac<strong>to</strong>rs, such as pregnancy,premature birth, chronic alcoholism, neoplasia, <strong>and</strong> cardiovascular disease. Themajority of isolations are from pregnant women, premature babies, <strong>and</strong> men <strong>and</strong>women over 45 years of age. The proportion of cases is higher in men than in women.Infection by C. fetus causes septicemia in man. In more than half of the cases,bacteremia is secondary <strong>and</strong> follows different localized infections. Between 17%<strong>and</strong> 43% of septicemic patients die (Morrison et al., 1990). Most cultures have beenobtained from the bloodstream during fever, but the etiologic agent has also beenisolated from synovial <strong>and</strong> spinal fluid, <strong>and</strong> sometimes from the feces of patientswith acute enteritis.In pregnant women, the illness has been observed from the fifth month of pregnancy,accompanied by a sustained fever <strong>and</strong> often by diarrhea. Pregnancy may terminatein miscarriage, premature birth, or full-term birth. Premature infants <strong>and</strong>some full-term infants die from the infection, which presents symp<strong>to</strong>ms of meningitisor meningoencephalitis. The syndrome may begin the day of birth with a slightfever, cough, <strong>and</strong> diarrhea; after two <strong>to</strong> seven days, the signs of meningitis appear.The case fatality rate is approximately 50%. Malnourished children, <strong>and</strong> at timesapparently healthy ones, can develop bacteremia along with vomiting, anorexia,diarrhea, <strong>and</strong> fever. The patient usually recovers spontaneously or after antibiotictreatment. In adults, often those already weakened by other illness, the diseaseappears as a generalized infection with extremely variable symp<strong>to</strong>ma<strong>to</strong>logy(Bokkenheuser <strong>and</strong> Sutter, 1981). C. fetus subsp. fetus is above all an opportunisticpathogen that gives rise <strong>to</strong> a systemic infection but rarely causes enteritis, in contrast<strong>to</strong> C. jejuni. Some cases of gastroenteritis caused by C. fetus subsp. fetus have beennoted in men without a compromised immune system (Devlin <strong>and</strong> McIntyre, 1983;Harvey <strong>and</strong> Greenwood, 1983).Gentamicin is the recommended antibiotic in the case of bacteremia <strong>and</strong> otherclinical forms of nonenteric infection. Chloramphenicol is recommended when thecentral nervous system is involved. Prolonged antibiotic treatment is necessary <strong>to</strong>prevent relapses (Morrison et al., 1990).The Disease in Animals: In cattle <strong>and</strong> sheep, vibriosis is an important diseasethat causes considerable losses due <strong>to</strong> infertility <strong>and</strong> abortions.CATTLE: In this species, the principal etiologic agent is C. fetus subsp. venerealis<strong>and</strong>, <strong>to</strong> a lesser degree, subsp. fetus. Genital vibriosis is a major cause of infertility,causing early embryonic death. The principal symp<strong>to</strong>m is the repetition of estrusafter service. During an outbreak, a high proportion of cows come in<strong>to</strong> heat repeat-


74 BACTERIOSESedly for three <strong>to</strong> five months, but only 25% <strong>to</strong> 40% of them become pregnant afterbeing bred twice. Of the cows or heifers that finally become pregnant, 5% <strong>to</strong> 10%abort five months in<strong>to</strong> gestation. An undetermined proportion of females harbor C.fetus subsp. venerealis during the entire gestation period <strong>and</strong> become a source ofinfection for the bulls in the next breeding season. After the initial infection, cowsacquire resistance <strong>to</strong> the disease <strong>and</strong> recover their normal fertility, i.e., the embryodevelops normally. However, immunity <strong>to</strong> the infection is only partial <strong>and</strong> the animalsmay become reinfected even though the embryos continue <strong>to</strong> develop normally.Resistance decreases substantially after three <strong>to</strong> four years.The infection is transmitted by natural breeding or artificial insemination. Bullsare the normal, though in most cases temporary, carriers of the infection. They playan important role in its transmission <strong>to</strong> females. The etiologic agent is carried in thepreputial cavity. Bulls may become infected while servicing infected cows, as wellas by contaminated instruments <strong>and</strong> equipment used in artificial insemination. Theetiologic agent is sensitive <strong>to</strong> antibiotics that are added <strong>to</strong> the semen used in artificialinsemination.C. fetus subsp. fetus is responsible for sporadic abortions in cattle. Some femalesare carriers of the infection, house the infectious agent in the gallbladder, <strong>and</strong> eliminateit in fecal matter.SHEEP: The principal agents of epizootic abortion in sheep are C. fetus subsp. fetus<strong>and</strong> C. jejuni <strong>and</strong>, <strong>to</strong> a lesser extent, C. fetus subsp. venerealis. The disease is characterizedby fetal death <strong>and</strong> abortions in the final months of gestation, or by fulltermbirth of dead lambs or lambs that die shortly thereafter. The infection also givesrise <strong>to</strong> metritus <strong>and</strong> placentitis, both of which may result in septicemia <strong>and</strong> death ofthe ewe. Losses of 10% <strong>to</strong> 20% of the lambs <strong>and</strong> 5% of the ewes that abort are <strong>common</strong>.The rate of abortions varies <strong>and</strong> depends on the proportion of susceptible ewes.Infected animals acquire immunity. Ewes do not abort again for about three years.If the infection is recent in the flock, the abortion rate can be quite high, at times up<strong>to</strong> 70% of the pregnant ewes. The infection is transmitted orally; venereal transmissionapparently plays no role.Source of Infection <strong>and</strong> Mode of Transmission (Figure 9): The reservoir of C.fetus is animals, but it is not clear how man contracts the infection. It is presumedthat he can become infected by direct contact with infected animals, by ingestion ofcontaminated food (unpasteurized milk, raw liver) or water, by transplacental transmission,exposure during birth, or sexual contact. It should be noted, however, thatsome patients have denied any contact with animals or even with products of animalorigin. It is also suspected that the infection may be endogenous. The etiologic agentwould be an oral commensal parasite that could penetrate the bloodstream during adental extraction. Another hypothesis is that C. fetus could be harbored in the humanintestine without becoming evident until the host loses resistance due <strong>to</strong> some illness.It would then invade through the mucosa, causing a generalized infection. Insummary, the source <strong>and</strong> pathogenesis of C. fetus in man continue <strong>to</strong> be unknown(Morrison et al., 1990).The sources of infection in cattle are carrier bulls <strong>and</strong> also cows that remaininfected from one parturition <strong>to</strong> the next. The mode of transmission is sexual contact.For sheep, the source of infection is environmental contamination. The placentasof infected sheep that abort or even of those that give birth normally, as well as


CAMPYLOBACTERIOSIS 75Figure 9. Campylobacteriosis (Campylobacter fetus). Probable mode of transmission.1. Campylobacter fetus subsp. venerealisBullSexual contactCow2. Campylobacter fetus subsp. fetus (intestinalis)SheepPlacenta,fetuses, excretaEnvironmentalcontamination(pasture, water,objects)Oral routeSheepDirect contact?Contamination of food?ManPlacental route?ManNOTE: It is not known how the disease is transmitted <strong>to</strong> humans; transmission is assumed <strong>to</strong> occur through direct contact,contamination of foods, or transplacental passage.aborted fetuses <strong>and</strong> vaginal discharges, contain a large number of Campylobacter. Afew infected ewes become carriers by harboring the infection in the gallbladder <strong>and</strong>shedding the agent in fecal matter. Contaminated grass, <strong>to</strong>ols, <strong>and</strong> clothing are thevehicles of infection. Transmission is oral. Sexual transmission has not been demonstrated,but knowledge on this subject is inadequate.Role of Animals in the Epidemiology of the Disease: Animals are the naturalreservoir of C. fetus. The agent has been observed <strong>to</strong> lodge in the human gallbladder,but it is not known how often man may become a carrier <strong>and</strong> give rise <strong>to</strong> humanfoci of infection. It is probably an exceptional occurrence. The mechanism of transmissionfrom animals <strong>to</strong> humans is unclear.Diagnosis: So far, diagnosis of campylobacteriosis in man has been largely fortui<strong>to</strong>us,when C. fetus is discovered in hemocultures of patients in whom the etiologywas not suspected. During the febrile period, repeated blood samples should betaken for culture. In cases of meningitis, cultures of cerebrospinal fluid should also


76 BACTERIOSESbe made. For isolation from vaginal fluid, repeated cultures on antibiotic media arerecommended.In cattle, diagnosis of epizootic infertility is based on the his<strong>to</strong>ry of the herd, onthe culture of the preputial secretion <strong>and</strong> semen from bulls <strong>and</strong> of vaginal mucusfrom nonpregnant cows <strong>and</strong> heifers, <strong>and</strong> also on culture of fluid from the abomasum<strong>and</strong> from the liver of aborted fetuses. All samples should be cultured within sixhours of collection. The highest rate of isolation of C. fetus from the cervicovaginalmucus is obtained in the two days immediately before or after estrus.When the infection is suspected in a herd of beef cattle, bacteriologic examinationof the cervicovaginal mucus of about 20 heifers that were bred but remained barrenis recommended. Samples should be taken six months after the start of the breedingseason.A good diagnostic technique for herd infection, though not for individual infection,is the agglutination test using cervicovaginal mucus. Another test in use is indirecthemagglutination, also employing vaginal mucus. Immunofluorescence is nonspecificin cows, since C. fetus subsp. venerealis gives cross-reactions with C. fetussubsp. fetus.Individual diagnosis is difficult in bulls. An isolation obtained from the preputialsecretion is conclusive if the culture is positive but not if it is negative. It is acceptedthat before a bull is introduced in<strong>to</strong> an artificial insemination center, he must passfour consecutive bacteriological tests at one-week intervals or four immunofluorescencetests. An excellent test is <strong>to</strong> have him service virgin heifers <strong>and</strong> subsequentlyculture their cervicovaginal mucus.In sheep, diagnosis is carried out primarily by culture of fetal tissue, afterbirths,<strong>and</strong> vaginal fluid. Fluid from the abomasum <strong>and</strong> liver of the fetus is preferable forisolation.Control: The few facts available at present on the epidemiology of the humaninfection are insufficient <strong>to</strong> determine control measures.The best method for preventing epizootic infertility in cattle is <strong>to</strong> use semen frominfection-free bulls in artificial insemination. In herds where this procedure is notpractical, cows <strong>and</strong> heifers can be vaccinated annually some two or three monthsbefore breeding using commercial bacterins with an adjuvant. Several trials offerevidence that vaccination with bacterins can also eliminate the carrier state in bulls<strong>and</strong> cows. The curative properties of the vaccines provide a new perspective in control.Nevertheless, it must be borne in mind that while this method can reduce theinfection in bulls under range conditions, vaccination of infected animals will noteliminate the infection from the herd. In one experiment (Vázquez et al., 1983), C.fetus subsp. venerealis was isolated from 2 out of 10 artificially infected bulls fiveweeks after administration of the recommended two doses one month apart.In sheep, good control can be obtained based on the vaccination of females withboth monovalent (with the subsp. fetus) <strong>and</strong> bivalent (fetus <strong>and</strong> venerealis) bacterinswith adjuvants, although the combined product is preferable. In flocks where adultfemales have acquired natural immunity, good results have been obtained by vaccinatingonly the yearly replacement ewes. Proper sanitary management is important,especially such measures as immediate removal of fetuses <strong>and</strong> afterbirths, isolationof sheep that have aborted, <strong>and</strong> protection of water from contamination.


CAMPYLOBACTERIOSIS 77BibliographyAndrews, P.J., F.W. Frank. Comparison of four diagnostic tests for detection of bovine genitalvibriosis. J Am Vet Med Assoc 165:695–697, 1974.Bokkenheuser, V. Vibrio fetus infection in man. I. Ten new cases <strong>and</strong> some epidemiologicobservations. Am J Epidemiol 91:400–409, 1970.Bokkenheuser, V.D., V.L. Sutter. Campylobacter infections. In: Balows, A., W.Y. Hausler,Jr., eds. Bacterial Mycotic <strong>and</strong> Parasitic Infections. 6th ed. Washing<strong>to</strong>n, D.C.: AmericanPublic Health Association; 1981.Bouters, R., J. De Keyser, M. F<strong>and</strong>eplassche, A. Van Aert, E. Brone, P. Bonte. Vibrio fetusinfection in bulls. Curative <strong>and</strong> preventive vaccination. Brit Vet J 129:52–57, 1973.Bryner, J.H. Vibriosis due <strong>to</strong> Vibrio fetus. In: Hubbert, W.T., W.F. McCulloch, P.R.Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield:Thomas; 1975.Bryner, J.H., P.C. Estes, J.W. Foley, P.A. O’Berry. Infectivity of three Vibrio fetus biotypesfor gallbladder <strong>and</strong> intestines of cattle, sheep, rabbits, guinea pigs, <strong>and</strong> mice. Am J Vet Res32:465–470, 1971.Bryner, J.H., P.A. O’Berry, A.H. Frank. Vibrio infection of the digestive organs of cattle.Am J Vet Res 25:1048–1050, 1964.Carroll, E.J., A.B. Hoerlein. Diagnosis <strong>and</strong> control of bovine genital vibriosis. J Am Vet MedAssoc 161:1359–1364, 1972.Clark, B.L. Review of bovine vibriosis. Aust Vet J 47:103–107, 1971.Clark, B.L., J.H. Duffy, M.J. Monsbourgh, I.M. Parsonson. Studies on venereal transmissionof Campylobacter fetus by immunized bulls. Aust Vet J 51:531–532, 1975.Devlin, H.R., L. McIntyre. Campylobacter fetus subsp. fetus in homosexual males. J ClinMicrobiol 18:999–1000, 1983.Firehammer, B.D., W.W. Hawkings. The pathogenicity of Vibrio fetus isolated from ovinebile. Cornell Vet 54:308–314, 1964.Harvey, S.M., J.R. Greenwood. Probable Campylobacter fetus subsp. fetus gastroenteritis.J Clin Microbiol 18:1278–1279, 1983.Hoerlein, A.B. Bovine genital vibriosis. In: Faulkner, L.C., ed. Abortion Diseases ofLives<strong>to</strong>ck. Springfield: Thomas; 1968.Hoerlein, A.B., E.J. Carroll. Duration of immunity <strong>to</strong> bovine genital vibriosis. J Am Vet MedAssoc 156:775–778, 1970.Laing, J.A. Vibrio fetus Infection of Cattle. Rome: Food <strong>and</strong> Agriculture Organization;1960. (Agricultural Studies 51).Miller, V.A. Ovine genital vibriosis. In: Faulkner, L.C., ed. Abortion Diseases of Lives<strong>to</strong>ck.Springfield: Thomas; 1968.Miner, M.L., J.L. Thorne. Studies on the indirect transmission of Vibrio fetus infection insheep. Am J Vet Res 25:474–477, 1964.Morrison, V.A., B.K. Lloyd, J.K.S. Chia, C.U. Tuazon. Cardiovascular <strong>and</strong> bacteremicmanifestations of Campylobacter fetus infection: Case report <strong>and</strong> review. Rev Infect Dis12:387–392, 1990.Osburn, B.I., R.K. Hoskins. Experimentally induced Vibrio fetus var. intestinalis infectionin pregnant cows. Am J Vet Res 31:1733–1741, 1970.Schurig, G.G.D., C.E. Hall, K. Burda, L.B. Corbeil, J.R. Duncan, A.J. Winter. Infection patternsin heifers following cervicovaginal or intrauterine instillation of Campylobacter (Vibrio)fetus venerealis. Cornell Vet 64:533–548, 1974.Schurig, G.G.D., C.E. Hall, L.B. Corbeil, J.R. Duncan, A.J. Winter. Bovine venerealvibriosis. Cure genital infection in females by systemic immunization. Infect Immun 11:245–251, 1975.S<strong>to</strong>rz, J., M.L. Miner, A.E. Olson, M.E. Marriott, Y.Y. Elsner. Prevention of ovine vibriosis byvaccination: Effect of yearly vaccination of replacement of ewes. Am J Vet Res 27:115–120, 1966.


78 BACTERIOSESVásquez, L.A., L. Ball, B.W. Bennett, G.P. Rupp, R. Ellis, J.D. Olson, et al. Bovine genitalcampylobacteriosis (vibriosis): Vaccination of experimentally infected bulls. Am J Vet Res44:1553–1557, 1983.Véron, M., R. Chatelain. Taxonomic study of the genus Campylobacter Sebald <strong>and</strong> Véron<strong>and</strong> designation of the neotype strain for the type species, Campylobacter fetus (Smith <strong>and</strong>Taylor) Sebald <strong>and</strong> Véron. Int J Syst Bacteriol 23:122–134, 1973.White, F.H., A.F. Walsh. Biochemical <strong>and</strong> serologic relationships of isolants of Vibrio fetusfrom man. J Infect Dis 121:471–474, 1970.CAT-SCRATCH DISEASEICD-10 A28.1Synonyms: Cat-scratch fever, benign inoculation lymphoreticulosis, cat-scratchsyndrome.Etiology: For many years, microbiologists were unable <strong>to</strong> identify the etiologicagent. Various microbes considered the etiologic agent at one time or another wereisolated; these included viruses, chlamydiae, <strong>and</strong> various types of bacteria. In 1983,Wear et al. conducted a his<strong>to</strong>pathologic examination of the lymph nodes of 39patients <strong>and</strong> demonstrated in 34 of them the presence of small, gram-negative, pleomorphicbacilli located in capillary walls or near areas of follicular hyperplasia <strong>and</strong>inside microabscesses. The observed bacilli were intracellular in the affected areas;they increased in number as lesions developed <strong>and</strong> diminished as they disappeared.The sera of three convalescent patients <strong>and</strong> human anti-immunoglobulin conjugatedwith peroxidase resulted in a precipitate with bacilli from the his<strong>to</strong>logical sectionsof different patients, demonstrating that they were serologically related (Wear et al.,1983). This finding was later confirmed by other researchers during the period1984–1986 in skin lesions, lymph nodes, <strong>and</strong> conjunctiva.Researchers managed <strong>to</strong> culture <strong>and</strong> isolate the bacillus in a biphasic medium ofbrain-heart infusion broth, as well as in tissue cultures (English et al., 1988;Birkness et al., 1992). It is a bacillus that is difficult <strong>to</strong> isolate <strong>and</strong> its dimensions areat the light microscope’s limit of resolution. A polar flagellum could be seen in electronmicroscope images. Depending on the temperature at which cultures are incubated,vegetative forms (at 32°C) or forms with defective walls (at 37°C) are seen.There are more vegetative bacilli in lesions of the skin <strong>and</strong> conjunctiva (at 32°C),<strong>and</strong> fewer in lymph node lesions (37°C). This would also explain why cat-scratchdisease (CSD) could only be reproduced in armadillos <strong>and</strong> not in guinea pigs <strong>and</strong>other <strong>common</strong> labora<strong>to</strong>ry animals.This bacillus, for which the name Afipia felis was suggested (Birkness et al.,1992), satisfies Koch’s postulates for being the etiologic agent of CSD, according <strong>to</strong>English et al. (1988). Birkness et al. were very cautious about considering A. felisthe etiologic agent of CSD. This caution appears <strong>to</strong> be well-founded, as a microor-


CAT-SCRATCH DISEASE 79ganism belonging <strong>to</strong> the rickettsiae, Bar<strong>to</strong>nella (formerly Rochalimaea) henselae,was recently detected, which could be the agent responsible for most cases of catscratchdisease <strong>and</strong> which also causes other <strong>diseases</strong> in man (see Infections causedby Rochalimaea henselae, in Volume 2: Chlamydioses, Rickettsioses, <strong>and</strong> Viroses).Geographic Distribution: Worldwide (Benenson, 1990). It occurs sporadically.According <strong>to</strong> Heroman <strong>and</strong> McCurley (1982), more than 2,000 cases occur each year.Approximately 75% of the cases occurred in children. Small epidemic outbreaks <strong>and</strong>familial clustering have been reported in several countries. When an outbreak occursin a family, there are usually several familial contacts in whom intracutaneous testswill be positive <strong>to</strong> the Hanger-Rose antigen. It is possible, but questionable, that severalendemic areas exist around Toron<strong>to</strong> (Canada), New York City (USA), <strong>and</strong>Alfortville (France). Positive intracutaneous tests have been obtained in 10% of thepopulation living in the vicinity of Alfortville, a result that is difficult <strong>to</strong> interpret.The Disease in Man: Seven <strong>to</strong> twenty days or more can elapse between the catscratch or bite (or other lesion caused by some inanimate object) <strong>and</strong> the appearanceof symp<strong>to</strong>ms. The disease is characterized by a regional lymphadenopathy withoutlymphangitis. In about 50% of the cases, primary lesions are seen at the point ofinoculation. These consist of partially healed ulcers surrounded by an erythema<strong>to</strong>usarea, or of erythema<strong>to</strong>us papules, pustules, or vesicles. Lymphadenitis is generallyunilateral <strong>and</strong> <strong>common</strong>ly appears in the epitrochlear, axillary, or cervical lymphnodes, or in the femoral <strong>and</strong> inguinal lymph gl<strong>and</strong>s. Swelling in the lymph gl<strong>and</strong>s,which is generally painful <strong>and</strong> suppurates in about 25% of patients, persists for periodsranging from a few weeks <strong>to</strong> a few months. A high proportion of patients showsigns of systemic infection, which consist of a low, short-lived fever <strong>and</strong>, less frequently,chills, anorexia, malaise, generalized pain, vomiting, <strong>and</strong> s<strong>to</strong>mach cramps.Morbilliform cutaneous eruptions sometimes occur.In general, the disease is benign <strong>and</strong> heals spontaneously without sequelae.Complications have been observed in a small proportion of the patients. The most<strong>common</strong> is Parinaud’s oculogl<strong>and</strong>ular syndrome; encephalitis, osteolytic lesions,<strong>and</strong> thrombocy<strong>to</strong>penic purpura are less frequent. The lymph gl<strong>and</strong> lesions are notpathognomonic, but they follow a certain pattern, which helps in diagnosis.His<strong>to</strong>pathologic studies have shown that alterations begin with hyperplasia of thereticular cells, followed by an inflamma<strong>to</strong>ry granuloma<strong>to</strong>us lesion. The center of thegranuloma degenerates <strong>and</strong> becomes a homogenous eosinophilic mass, in whichabscesses <strong>and</strong> microabscesses later appear.In a study of 76 cases with neurological complications (51 with encephalopathy<strong>and</strong> 15 with disorders of the cranial or peripheral nerves), 50% of the patients had afever, but only 26% had temperatures above 30°C. Forty-six percent of the patientshad convulsions <strong>and</strong> 40% displayed aggressive behavior. Lethargy, with or withoutcoma, was accompanied by various neurological symp<strong>to</strong>ms. Of the other 15 patientswithout encephalopathy, 10 had neuroretinitis, two children had facial paresis, <strong>and</strong>three women had peripheral neuritis. Seventy-eight percent of the patients recoveredwithout sequelae within a period of 1 <strong>to</strong> 12 weeks <strong>and</strong> the rest recovered within ayear. Treatment consisted of controlling the convulsions <strong>and</strong> support measures.Commonly used antibiotics were apparently ineffective (Carithers <strong>and</strong> Margileth,1991). Infection of the viscera is rare, but has been reported as well (Delahoussaye<strong>and</strong> Osborne, 1990).


80 BACTERIOSESMost cases have occurred in children, who have more contact with cats.In temperate climates, the disease tends <strong>to</strong> be seasonal, with most cases occurringin fall <strong>and</strong> winter. In hot climates, there are no seasonal differences.Source of Infection <strong>and</strong> Mode of Transmission: The most salient fact in the epidemiologyof this disease is its causal relation with a cat scratch. It is estimated thatabout 65% of patients were scratched or bitten by cats <strong>and</strong> that 90% of the cases hadsome contact with these animals. Nevertheless, cases have been observed in whichthe skin lesion was inflicted by such inanimate objects as splinters, thorns, or pins.Cats undoubtedly play an important role in the epidemiology, but there is doubtabout whether it is as host for the etiologic agent or simply as a mechanical vec<strong>to</strong>r.Another possibility is that the etiologic agent is part of the normal flora of the cat’smouth <strong>and</strong> is transferred <strong>to</strong> the nails when the cat grooms itself (Hainer, 1987).Several observations—among them the fact that some cases were caused by inanimateagents—suggest that cats could be mechanical transmitters. Cats implicated inhuman cases were healthy animals, almost always young, that did not react <strong>to</strong> theHanger-Rose intradermal test. It is also interesting <strong>to</strong> note that cats inoculated withmaterial from the lymph nodes of human patients did not become ill. In summary, ithas not yet been possible <strong>to</strong> show that cats are infected with the disease or are carriersof its causal agent, despite the many attempts made. According <strong>to</strong> Margileth(1987), cats are only able <strong>to</strong> transmit the infection for a short time (two <strong>to</strong> threeweeks). CSD is usually transmitted from cats <strong>to</strong> man through a scratch <strong>and</strong>, less frequently,through a bite or licking. In Parinaud’s oculogl<strong>and</strong>ular syndrome, the poin<strong>to</strong>f entry for the agent is the conjunctiva or eyelids when a person rubs his or her eyesafter picking up a cat (August, 1988).Diagnosis: CSD can be clinically confused with other <strong>diseases</strong> that cause regionallymphadenopathies, such as tularemia, brucellosis, tuberculosis, pasteurellosis,infectious mononucleosis, Hodgkin’s disease, venereal lymphogranuloma, lymphosarcoma,<strong>and</strong> lymphoma. All these <strong>diseases</strong> must be excluded before consideringa diagnosis of CSD. The symp<strong>to</strong>ms described above, a his<strong>to</strong>ry of a skin lesioncaused by a cat scratch or bite, the his<strong>to</strong>pathology of biopsy material taken from theaffected lymph node, <strong>and</strong> the Hanger-Rose intradermal test constitute the basis fordiagnosis. The Hanger-Rose antigen is prepared by suspending pus taken from anabscessed lymph node in a 1:5 saline solution <strong>and</strong> heating it for 10 hours at 60°C.The antigen is very crude <strong>and</strong> difficult <strong>to</strong> st<strong>and</strong>ardize. The test is carried out by intradermalinoculation with 0.1 ml of the antigen. The reaction may be read in 48 hours.Edema measuring 0.5 cm <strong>and</strong> erythema of 1 cm are considered a positive reaction.The test is very useful, since 90% of 485 clinically diagnosed cases gave positiveresults, while only 4.1% out of 591 controls tested positive.There is a danger of transmitting viral hepatitis with this antigen; therefore, thepreparation should be heat-treated for a lengthy period, as indicated above. It maybe very useful <strong>to</strong> demonstrate the presence of the putative etiologic agent, A. felis,using Warthin-Starry stain on his<strong>to</strong>logical sections from the skin or lymph nodes.Control: Prevention is limited <strong>to</strong> avoiding cat scratches <strong>and</strong> bites. Cutting thecat’s nails, washing <strong>and</strong> disinfecting any scratch or bite, <strong>and</strong> washing one’s h<strong>and</strong>safter petting or h<strong>and</strong>ling a cat are also recommended (August, 1988).


CAT-SCRATCH DISEASE 81BibliographyAndrews, C., H.G. Pereira. Viruses of Vertebrates. 3rd ed. Baltimore: Williams & Wilkins; 1972.August, J.R. Cat-scratch disease. Zoonosis update. J Am Vet Med Assoc 193:312–315, 1988.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Birkness, K.A., V.G. George, E.H. White, et al. Intracellular growth of Afipia felis, a putativeetiologic agent of cat-scratch disease. Infect Immun 60:2281–2287, 1992.Carithers, H.A. Cat-scratch disease. An overview based on a study of 1,200 patients. Am JDis Child 139:1124–1133, 1985.Carithers, H.A., A.M. Margileth. Cat-scratch disease. Acute encephalopathy <strong>and</strong> other neurologicmanifestations. Am J Dis Child 145:98–101, 1991.Delahoussaye, P.M., B.M. Osborne. Cat-scratch disease presenting as abdominal visceralgranulomas. J Infect Dis 161:71–78, 1990.Emmons, R.W., J.L. Riggs, J. Schachter. Continuing search for the etiology of cat-scratchdisease. J Clin Microbiol 4:112–114, 1976.English, C.K, D.J. Wear, A.M. Margileth, et al. Cat-scratch disease. Isolation <strong>and</strong> culture ofthe bacterial agent. JAMA 259:1347–1352, 1988.Euseby, J.B. Le genre Afipia et la maladie de griffes du chat. Revue Med Vet 143:95–105,1992.Gerber, M.A., A.K. Sedgwick, T.J. MacAlister, K.B. Gustafson, M. Ballow, R.C. Til<strong>to</strong>n.The aetiological agent of cat-scratch disease. Lancet 1:1236–1239, 1985.Griesemer, R.A., L.G. Wolfe. Cat-scratch disease. J Am Vet Med Assoc 158:1008–1012, 1971.Hainer, B.L. Cat-scratch disease. J Fam Pract 25:497–503, 1987.Heroman, V.M, W.S. McCurley. Cat-scratch disease. O<strong>to</strong>laryngol Clin North Am15:649–658, 1982. Cited in: Delahoussaye, P.M., B. Osborne. Cat-scratch disease presentingas abdominal visceral granulomas. J Infect Dis 161:71–78, 1990.Macrae, A.D. Cat-scratch fever. In: Graham-Jones, O., ed. Some Diseases of AnimalsCommunicable <strong>to</strong> Man in Britain. Oxford: Pergamon Press; 1968.Margileth, A.M. Cat-scratch disease. A therapeutic dilemma. Vet Clin North Am Small AnimPract 17:91–103, 1987. Cited in: August, J.R. Cat-scratch disease. Zoonosis update. J Am VetMed Assoc 193:312–315, 1988.Rose, H.M. Cat-scratch disease. In:Wyngaarden, J.B., L.H. Smith, Jr., eds. Cecil Textbookof Medicine. 16th ed. Philadelphia: Saunders; 1982.Warwick, W.J. The cat-scratch syndrome, many <strong>diseases</strong> or one disease. Progr Med Virol9:256–301, 1967.Wear, D.J., A.M. Margileth, T.L. Hadfield, et al. Cat-scratch disease: A bacterial infection.Science 221:1403–1404, 1983.


82 BACTERIOSESCLOSTRIDIAL FOOD POISONINGICD-10 A05.2 foodborne Clostridium perfringens[Clostridium welchii] in<strong>to</strong>xicationSynonyms: Clostridial gastroenteritis, clostridial <strong>to</strong>xicosis.Etiology: Clostridium perfringens (C. welchii) is an anaerobic, gram-positive,sporogenic, nonmotile, encapsulated bacillus that produces extracellular <strong>to</strong>xins. Theoptimum temperature for its growth is between 41°C <strong>and</strong> 45°C. At these temperatures,C. perfringens reproduces at what is considered record speed for most bacteria.This growth potential is very important in food protection. A temperature of 60°Cis lethal for the vegetative form of C. perfringens in culture media. It is more resistant<strong>to</strong> heat when found in foods (Labbe, 1989). Five different <strong>to</strong>xigenic types areknown, designated by the letters A through E; these produce four principal <strong>to</strong>xins.The vegetative forms produce large quantities of entero<strong>to</strong>xins during sporulation inthe intestine. The optimum temperature for sporulation is between 35°C <strong>and</strong> 40°C.Geographic Distribution: C. perfringens type A is ubiqui<strong>to</strong>us in the soil <strong>and</strong> inthe intestinal tract of humans <strong>and</strong> animals worldwide. The other types are found onlyin the intestinal tract of animals. Types B <strong>and</strong> E have a marked regional distribution.Occurrence in Man: Outbreaks of food poisoning due <strong>to</strong> C. perfringens type Aprobably occur the world over, but most of the information comes from developedcountries.In Great Britain, where food poisoning is a notifiable disease, clostridial poisoningis estimated <strong>to</strong> cause 30% of all cases, as well as many general <strong>and</strong> familial outbreaks;an average of 37 people are affected per outbreak.In the United States, during the period 1976–1980, 62 outbreaks affecting 6,093persons were reported, representing 7.4% of all outbreaks of food <strong>to</strong>xicoses withknown etiology <strong>and</strong> 14.8% of the <strong>to</strong>tal number of known cases in the country overthe same period. The median number of cases per outbreak was 23.5, but six outbreaksaffected more than 200 persons (Sh<strong>and</strong>era et al., 1983).Even in developed countries, cases are greatly underreported, because the diseaseis mild <strong>and</strong> usually lasts no more than 24 hours. Moreover, labora<strong>to</strong>ry diagnosis cannotalways be performed, as it depends on obtaining food <strong>and</strong> patient s<strong>to</strong>ol samplesthat are not always available.Outbreaks affecting large numbers of people are usually reported. These arecaused by meals prepared in restaurants or institutions. An outbreak occurred inArgentina at the farewell ceremony for 60 participants in an international course.The food served included meat pies, canapés, <strong>and</strong> sweet cakes provided by a restaurant.Of the 41 people who were still in the country during the epidemiologicalinvestigation, 56% reported having developed symp<strong>to</strong>ms typical of gastroenteritis.The meat pies were considered the source of the poisoning (Michanie et al., 1993).In New Guinea, necrotic enteritis in man caused by C. perfringens type C hasbeen confirmed.Occurrence in Animals: In domesticated ruminants, several types of extero<strong>to</strong>xemiasdue <strong>to</strong> C. perfringens types B, C, D, <strong>and</strong> E are known. Entero<strong>to</strong>xemia results


CLOSTRIDIAL FOOD POISONING 83from the absorption in<strong>to</strong> the bloodstream of <strong>to</strong>xins produced in the intestine by thevarious types of C. perfringens that form part of the normal intestinal flora.The Disease in Man: The disease is contracted upon ingestion of foods (especiallyred meat <strong>and</strong> fowl) in which C. perfringens type A has multiplied. It is nowknown that illness is caused by thermoresistant strains, which can survive at 100°Cfor more than an hour, as well as by thermolabile <strong>and</strong> hemolytic strains, which areinactivated after approximately 10 minutes at 100°C.The incubation period is from 6 <strong>to</strong> 24 hours after ingestion, but has been as shortas two hours in a few people, which indicates that the food ingested contained preformed<strong>to</strong>xin. The disease begins suddenly, causing abdominal cramps <strong>and</strong> diarrhea,but usually not vomiting or fever. It lasts a day or less <strong>and</strong> its course is benign,except in debilitated persons, in whom it may prove fatal. Food poisoning caused byC. perfringens type A does not usually require medical treatment.In recent years, an intestinal infection with diarrhea not associated with food consumptionhas been described. The disease is due <strong>to</strong> an infection caused by colonizationof C. perfringens in the intestine <strong>and</strong> the production of entero<strong>to</strong>xin. Its clinicalpicture is very different from that of clostridial food poisoning <strong>and</strong> more closelyresembles an infection caused by Salmonella or Campylobacter. In Engl<strong>and</strong>, a seriesof cases was described involving 50 elderly patients (ages 76 <strong>to</strong> 96) who were hospitalizedwith diarrhea not associated with food consumption. The diarrhea lastedfor an average of 11 days, but lasted for a shorter period in two-thirds of the patients.Sixteen of 46 patients had bloody s<strong>to</strong>ols (Larson <strong>and</strong> Borriello, 1988).Necrotic enteritis produced by the ingestion of food contaminated with C. perfringenstype C is characterized by a regional gangrene in the small intestine, especiallythe jejunum.A rare type of necrotizing enteritis caused by C. perfringens type A was describedin the Netherl<strong>and</strong>s in a 17-year-old girl. The patient recovered after resection ofthree meters of her intestine <strong>and</strong> intravenous treatment with gentamicin, cefotaxime,<strong>and</strong> metronidazole for seven days. Counterimmunoelectrophoresis of blood samplesindicated the presence of antibodies for the alpha <strong>to</strong>xin that is predominant in typeA. A similar illness appeared in Germany <strong>and</strong> Norway after the Second World War.Currently, necrotic enteritis is rare in the Western world, though some cases amongadolescents <strong>and</strong> the elderly have been described (Van Kessel et al., 1985).On rare occasions, gastroenteritis due <strong>to</strong> C. perfringens type D has been confirmedin man. This type causes entero<strong>to</strong>xemia in sheep <strong>and</strong> goats.The Disease in Animals: C. perfringens type A is part of the normal flora of theintestine, where it does not usually produce its characteristic alpha <strong>to</strong>xin. Few casesof illness caused by type A have been confirmed in cattle. In California <strong>and</strong> Oregon(USA), a disease produced by type A in nursing lambs (“yellow lamb disease”) hasbeen described. The disease occurs in spring, when there is a large population ofnursing animals. The lambs suffer depression, anemia, jaundice, <strong>and</strong> hemoglobinuria.They die 6 <strong>to</strong> 12 hours after the onset of clinical symp<strong>to</strong>ms (Gillespie <strong>and</strong>Timoney, 1981).Type B is the etiologic agent of “lamb dysentery,” which occurs in Great Britain,the Middle East, <strong>and</strong> South Africa. It usually attacks lambs less than 2 weeks old. Itis characterized by hemorrhagic enteritis, <strong>and</strong> is frequently accompanied by ulcerationof the mucosa. It also affects calves <strong>and</strong> colts.


84 BACTERIOSESType C causes hemorrhagic entero<strong>to</strong>xemia (“struck”) in adult sheep in GreatBritain, as well as necrotic enteritis in calves, lambs, suckling pigs, <strong>and</strong> fowl inmany parts of the world (Timoney et al., 1988).Type D is the causal agent of entero<strong>to</strong>xemia in sheep. It is distributed worldwide<strong>and</strong> attacks animals of all ages. The disease is associated with abundant consumptionof food, whether milk, pasture, or grains. Outbreaks have also been describedin goats <strong>and</strong>, more rarely, in cattle.Type E causes dysentery or entero<strong>to</strong>xemia in calves <strong>and</strong> lambs, <strong>and</strong> has been confirmedin the US, Engl<strong>and</strong>, <strong>and</strong> Australia (Timoney et al., 1988).Source of Infection <strong>and</strong> Mode of Transmission: The natural reservoir of C. perfringenstype A is the soil <strong>and</strong> the intestine of man <strong>and</strong> animals. Some studies(Torres-Anjel et al., 1977) have shown that man harbors higher numbers of C. perfringensthan fowl or cattle <strong>and</strong> that some people excrete great quantities of thesebacteria, making man the most important reservoir of clostridial food poisoning. Theamount of C. perfringens type A in the intestine varies with the animal species <strong>and</strong>location. C. perfringens is found in large numbers in the small intestine of pigs, insmall amounts in sheep, goats, <strong>and</strong> cattle, <strong>and</strong> is practically nonexistent in horses(Smith, 1965).Type A entero<strong>to</strong>xemia is caused primarily by the alpha <strong>to</strong>xin, which forms in theintestine <strong>and</strong> is released during sporulation, for which the small intestine is a favorableenvironment. The source of poisoning for man is food contaminated by sporesthat survive cooking. Heat (heat shock) activates the spores, which then germinate.The vegetative forms multiply rapidly if the prepared food is left at room temperature,<strong>and</strong> can reach very high concentrations if the temperature is high for a sufficientamount of time (see the section on etiology). The vegetative forms carried <strong>to</strong>the intestine by the food sporulate, releasing the entero<strong>to</strong>xin in the process. The foodvehicle is almost always red meat or fowl, since they provide C. perfringens with theamino acids <strong>and</strong> vitamins it needs. Less frequently, other foods, such as pigeon peas,beans, mashed pota<strong>to</strong>es, cheeses, seafood, pota<strong>to</strong> salad, noodles, <strong>and</strong> olives havegiven rise <strong>to</strong> the disease (Craven, 1980). Immersing meat in broth or cooking it inlarge pieces creates anaerobic conditions that favor the multiplication of the bacteriaduring cooling or s<strong>to</strong>rage. The foods that cause poisoning are usually prepared inlarge quantities by restaurants or dining halls <strong>and</strong> are served later that day or thenext. The spores of some strains of C. perfringens can be destroyed by adequatecooking, but other spores are heat-resistant. Reheating food before serving it canstimulate the multiplication of bacteria if the heating temperature is not highenough. It is now known that high concentrations of the vegetative form of C. perfringensin food cannot be destroyed by s<strong>to</strong>mach acid, <strong>and</strong> thus pass in<strong>to</strong> the intestine.The entero<strong>to</strong>xin synthesized in the intestine when the bacteria sporulate isresistant <strong>to</strong> intestinal enzymes, has a cy<strong>to</strong><strong>to</strong>xic effect on the intestinal epithelium,affects the electrolyte transport system, <strong>and</strong> thus causes diarrhea (Narayan, 1982).It should be borne in mind that not all strains of C. perfringens are <strong>to</strong>xigenic. Onestudy of strains implicated in food poisonings found that 86% were <strong>to</strong>xigenic, whileanother study found that 2 strains out of 174 isolated from other sources producedthe entero<strong>to</strong>xin (Narayan, 1982).In lamb dysentery caused by C. perfringens type B, the animals are infected duringthe first days of life, apparently from the mother or the environment. Young


CLOSTRIDIAL FOOD POISONING 85lambs that receive a lot of milk are particularly likely <strong>to</strong> fall ill. The bacteria multiply<strong>and</strong> produce beta <strong>to</strong>xin when they sporulate (Timoney et al., 1988).In hemorrhagic enteritis or “struck” caused by C. perfringens type C in adult sheepin Engl<strong>and</strong>, the agent is found in the soil of areas of Romney Marsh <strong>and</strong> it is possiblethat most of the sheep in the region are infected. Beta <strong>to</strong>xin predominates. The soil <strong>and</strong>the intestinal tract of healthy sheep are the reservoir for type D, which is the agent ofentero<strong>to</strong>xemia in sheep. Epsilon <strong>to</strong>xin is the most important (Timoney et al., 1988).The intestines of 75 animals with diarrhea of unknown origin were examinedpostmortem <strong>to</strong> detect the presence of C. perfringens entero<strong>to</strong>xins. Positive resultswere found in 8 of 37 swine, 4 of 10 sheep, 1 of 3 goats, 1 of 16 cattle, <strong>and</strong> none of9 horses (Van Baelen <strong>and</strong> Devriese, 1987).In animals, C. perfringens seems <strong>to</strong> multiply primarily in the intestine, where itsporulates <strong>and</strong> produces <strong>to</strong>xins. The types of C. perfringens (B, C, D, E) that produceentero<strong>to</strong>xemia in animals multiply rapidly in the intestine <strong>and</strong> produce <strong>to</strong>xinswhen animals are suddenly released <strong>to</strong> rich pastures, are given <strong>to</strong>o much fodder, orconsume large quantities of milk.Role of Animals in the Epidemiology of the Disease: Human food poisoning iscaused by foods contaminated by C. perfringens type A, usually foods consistingmainly of red meat or fowl. The animals themselves do not play a direct role in theepidemiology, since the etiologic agent is ubiqui<strong>to</strong>us <strong>and</strong> can be found in the soil orin dust. Foods of animal origin are important as substrates for the multiplication ofthe bacteria <strong>and</strong> as vehicles for the disease. The soil <strong>and</strong> the intestines of humans<strong>and</strong> animals are the reservoir of the etiologic agent. C. perfringens type A is foundin the muscles <strong>and</strong> organs of animals a few hours after slaughter, unless they are rapidlyrefrigerated.Heat-resistant strains of C. perfringens may be found in the mesenteric lymphnodes of some animals after slaughter. Strains are isolated at a lower rate in animalsallowed <strong>to</strong> rest 24 <strong>to</strong> 48 hours before butchering.Diagnosis: The incubation period <strong>and</strong> clinical picture make it possible <strong>to</strong> distinguishclostridial food poisoning, which is afebrile, from salmonellosis, shigellosis,or colibacillosis, which produce fever. Staphylococcal in<strong>to</strong>xication usually results invomiting, while this symp<strong>to</strong>m is rare with clostridial poisoning. Labora<strong>to</strong>ry confirmationis based on the C. perfringens count in the implicated food <strong>and</strong> in thepatient’s s<strong>to</strong>ol (within 48 hours of onset of illness). The existence of 10 5 cells pergram of food <strong>and</strong> 10 6 per gram of fecal material is considered significant. Serotypingof strains from food <strong>and</strong> feces with a battery of 70 sera has provided good results inepidemiological research in Great Britain, but not in the United States, where only40% of the strains received by the Centers for Disease Control <strong>and</strong> Prevention couldbe typed. There is no proof that only certain serotypes are related <strong>to</strong> the disease(Sh<strong>and</strong>era, 1983).Labora<strong>to</strong>ry diagnosis of animal entero<strong>to</strong>xemias is performed by mouse inoculation<strong>to</strong> demonstrate the presence of specific <strong>to</strong>xins. Some mice are inoculated onlywith intestinal contents <strong>and</strong> others are inoculated with both intestinal contents <strong>and</strong>anti<strong>to</strong>xin. Tests <strong>to</strong> directly detect the <strong>to</strong>xin can also be performed <strong>and</strong> are currentlypreferred. These include reverse passive latex agglutination, enzyme immunoassay,or culturing of Vero cells with neutralizing antibodies <strong>to</strong> inhibit the cy<strong>to</strong>pathiceffects (Bartlett, 1990).


86 BACTERIOSESControl: In man, the control measures are as follows. Meat dishes should beserved hot <strong>and</strong> as soon as possible after cooking. If food must be kept for a whilebefore eating, it should be rapidly refrigerated. If possible, meat should be cut in<strong>to</strong>small pieces for cooking. Broth should be separated from the meat. The use of pressurecookers is a good preventive measure. If necessary, food should be reheated ata temperature high enough <strong>to</strong> destroy the agent’s vegetative cells.Educating those who prepare meals in restaurants or at home is very important,since it is impossible <strong>to</strong> avoid the presence of C. perfringens in red meats <strong>and</strong> rawchicken (Michanie et al., 1993).In animals, entero<strong>to</strong>xemia control consists of good herd management, avoidanceof a sudden change from poor <strong>to</strong> rich pasture, <strong>and</strong> active immunization with specific<strong>to</strong>xoids. Two doses of <strong>to</strong>xoid a month apart, followed by a booster at six months(type D) or a year (type C), are recommended.To protect lambs, ewes should be vaccinated with two doses, with the second doseadministered two weeks before lambing. To prevent lamb dysentery (type B), ewescan be vaccinated with the specific <strong>to</strong>xoid or lambs can be passively immunizedwith antiserum at birth. In C. perfringens types B <strong>and</strong> C, the beta <strong>to</strong>xin predominates,<strong>and</strong> therefore a <strong>to</strong>xoid or antiserum from one type will give cross-immunity.BibliographyBartlett, J.G. Gas gangrene (other clostridium-associated <strong>diseases</strong>). In: M<strong>and</strong>ell, G.L., R.G.Douglas, Jr., J.E. Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. NewYork: Churchill Livings<strong>to</strong>ne, Inc.; 1990.Craven, S.E. Growth <strong>and</strong> sporulation of Clostridium perfringens in foods. Food Techn34:80–87, 1980.Dobosch, D., R. Dowell. Detección de entero<strong>to</strong>xina de Clostridium perfringens en casos dein<strong>to</strong>xicación alimentaria. Medicina 43:188–192, 1983.Faich, G.A., E.J. Gangarosa. Food poisoning, bacterial. In: Top, F.M., P.F. Wehrle, eds.Communicable <strong>and</strong> Infectious Diseases. 7th ed. Saint Louis: Mosby; 1972.Gillespie, J.H., J.F. Timoney. Hagan <strong>and</strong> Bruner’s Infectious Diseases of DomesticAnimals. 7th ed. Ithaca: Cornell University Press; 1981.Hobbs, B.C. Clostridium perfringens <strong>and</strong> Bacillus cereus infections. In: Riemann, H., ed.Food-borne Infections <strong>and</strong> In<strong>to</strong>xications. New York: Academic Press; 1969.Labbe, R. Clostridium perfringens. In: Doyle, M.P., ed. Food-borne Bacterial Pathogens.New York: Marcel Dekker; 1989.Larson, H.E., S.P. Borriello. Infectious diarrhea due <strong>to</strong> Clostridium perfringens. J Infect Dis157:390–391, 1988.Michanie, S., A. Vega, G. Padilla, A. Rea Nogales. Brote de gastroenteritis provocado porel consumo de empanadas de carne. Alimentacion Latinoamer 194:49–54, 1993.Narayan, K.G. Food-borne infection with Clostridium perfringens Type A. Int J Zoonoses9:12–32, 1982.Roberts, R.S. Clostridial <strong>diseases</strong>. In: Stableforth, A.W., I.A. Galloway, eds. InfectiousDiseases of Animals. London: Butterworths; 1959.Rose, H.M. Diseases caused by Clostridia. In:Wyngaarden, J.B., L.H. Smith, Jr., eds. CecilTextbook of Medicine. 16th ed. Philadelphia: Saunders; 1982.Sh<strong>and</strong>era, W.X., C.O. Tacket, P.A. Blake. Food poisoning due <strong>to</strong> Clostridium perfringensin the United States. J Infect Dis 147:167–170, 1983.Smith, H.W. Observations on the flora of the alimentary tract of animals <strong>and</strong> fac<strong>to</strong>rs affectingits composition. J Path Bact 89:95–122, 1965. Cited in: Van Baelen, D., L.A. Devriese.


CLOSTRIDIAL WOUND INFECTIONS 87Presence of Clostridium perfringens entero<strong>to</strong>xin in intestinal samples from farm animals withdiarrhoea of unknown origin. J Vet Med B 34:713–716, 1987.Smith, L.D.S. Clostridial <strong>diseases</strong> of animals. Adv Vet Sci 3:463–524, 1957.Timoney, J.F., J.H. Gillespie, F.W. Scott, J.E. Barlough. Hagan <strong>and</strong> Bruner’s Microbiology<strong>and</strong> Infectious Diseases of Domestic Animals. 8th ed. Ithaca: Coms<strong>to</strong>ck; 1988.Torres-Anjel, M.J., M.P. Riemann, C.C. Tsai. Entero<strong>to</strong>xigenic Clostridium perfringens TypeA in Selected Humans: A Prevalence Study. Washing<strong>to</strong>n, D.C.: Pan American HealthOrganization; 1977. (Scientific Publication 350).Van Baelen, D., L.A. Devriese. Presence of Clostridium perfringens entero<strong>to</strong>xin inintestinal samples from farm animals with diarrhoea of unknown origin. J Vet Med B34:713–716, 1987.Van Kessel, L.J.P., H.A. Verbugh, M.F. Stringer, et al. Necrotizing enteritis associated with<strong>to</strong>xigenic Type A Clostridium perfringens [letter]. J Infect Dis 151:974–975, 1985.CLOSTRIDIAL WOUND INFECTIONSICD-10 A48.0 gas gangreneSynonyms: Gas gangrene, clostridial myonecrosis, his<strong>to</strong><strong>to</strong>xic infection, anaerobiccellulitis; malignant edema (in animals).Etiology: Wound infection is characterized by mixed bacterial flora. The mostimportant species are Clostridium perfringens (welchii), C. novyi, C. septicum, C.sordelli, C. his<strong>to</strong>lyticum, <strong>and</strong> C. fallax. Like all clostridia, these bacteria are grampositive,anaerobic, sporogenic bacilli. These species produce potent exo<strong>to</strong>xins thatdestroy tissue. In human gas gangrene, the most important etiologic agent is C. perfringens,<strong>to</strong>xigenic type A. Infection by C. septicum predominates in animals.Geographic Distribution: Worldwide.Occurrence in Man: Gas gangrene used <strong>to</strong> be more prevalent in wartime than inpeacetime. It has been estimated that during World War I, 100,000 German soldiersdied from this infection. However, its incidence has decreased enormously duringmore recent wars. During the eight years of the Vietnam War, there were only 22cases of gas gangrene out of 139,000 wounds, while in Miami (USA), there were27 cases in civilian trauma patients over a 10-year period (Finegold, 1977). Thedisease is relatively rare <strong>and</strong> occurs mainly in traffic- <strong>and</strong> occupational accidentvictims. However, in natural disasters or other emergencies, it constitutes a seriousproblem. Gas gangrene also occurs after surgery, especially in older patients whohave had a leg amputated. It may also develop in patients receiving intramuscularinjections, especially of medications suspended in an oil base. Gas gangrene canoccur in soft tissue lesions in patients with vascular insufficiency, such as diabetics(Bartlett, 1990).Occurrence in Animals: The frequency of occurrence in animals is not known.


88 BACTERIOSESThe Disease in Man: Pathogenic species of Clostridium may be found as simplecontaminants in any type of traumatic lesion. When infection occurs, the microorganismsmultiply <strong>and</strong> produce gas in the tissues. Gas gangrene is an acute <strong>and</strong> seriouscondition that produces myositis as its principal lesion. The incubation periodlasts from six hours <strong>to</strong> three days after injury. The first symp<strong>to</strong>ms are increasing painaround the injured area, tachycardia, <strong>and</strong> decreased blood pressure, followed byfever, edematization, <strong>and</strong> a reddish serous exudate from the wound. The skinbecomes taut, discolored, <strong>and</strong> covered with vesicles. Crepitation is felt upon palpation.Stupor, delirium, <strong>and</strong> coma develop in the final stages of the disease. The infectionmay also begin in the uterus following an abortion or difficult labor. These casesshow septicemia, massive hemolysis, <strong>and</strong> acute nephrosis, with shock <strong>and</strong> anuria.C. perfringens type A, alone or in combination with other pathogens, caused 60%<strong>to</strong> 80% of gas gangrene cases in soldiers during the two world wars.Treatment consists primarily of debridement with extensive removal of theaffected muscle. Amputation of the limb affected by gas gangrene should be considered.Penicillin G is generally the preferred antibacterial. However, better resultshave been obtained with clindamycin, metronidazole, rifampicin, <strong>and</strong> tetracycline(Bartlett, 1990). Mortality is still very high.The Disease in Animals: C. septicum is the principal agent of clostridial woundinfection, known as “malignant edema.” C. septicum produces four <strong>to</strong>xins that causetissue damage. The incubation period lasts from a few hours <strong>to</strong> several days. This diseaseis characterized by an extensive hemorrhagic edema of the subcutaneous tissue<strong>and</strong> intermuscular connective tissue. The muscle tissue turns dark red; little or no gasis present. The infected animal exhibits fever, in<strong>to</strong>xication, <strong>and</strong> lameness. Swellingsare soft <strong>and</strong> palpation leaves depressions. The course of the disease is rapid <strong>and</strong> theanimal can die a few days after symp<strong>to</strong>ms appear. Cattle are the most affected species,but sheep, horses, <strong>and</strong> swine are also susceptible. The infection is rare in fowl.C. perfringens type A is sometimes responsible for infection of traumatic woundsin calves, lambs, <strong>and</strong> goats. As in man, the infection gives rise <strong>to</strong> gas gangrene.Edema with a large amount of gas develops around the injury site, spreads rapidly,<strong>and</strong> causes death in a short time.In animals, as in man, other clostridia (e.g., C. novyi, C. sordelli, <strong>and</strong> C. his<strong>to</strong>lyticum)can cause wound infection <strong>and</strong> the wound’s bacterial flora may be mixed.Treatment with high doses of penicillin or broad-spectrum antibiotics may yieldresults if administered at the onset of disease.Source of Infection <strong>and</strong> Mode of Transmission: Clostridia are widely distributedin nature, in the soil, <strong>and</strong> in the intestinal tract of man <strong>and</strong> most animals. Thesources of infection for man <strong>and</strong> animals are the soil <strong>and</strong> fecal matter. Transmissionis effected through traumatic wounds or surgical incisions. Gas gangrene can alsooccur without any wound or trauma (endogenous or spontaneous gas gangrene) inpatients weakened by malignant disease <strong>and</strong> those with ulcerative lesions in the gastrointestinalor urogenital tract or in the bile ducts (Finegold, 1977). In animals, theinfection may originate in minor wounds, such as those produced by castration, taildocking, <strong>and</strong> shearing.Role of Animals in the Epidemiology of the Disease: Wound clostridiosis is adisease <strong>common</strong> <strong>to</strong> man <strong>and</strong> animals, not a zoonosis.


CLOSTRIDIAL WOUND INFECTIONS 89Diagnosis: Diagnosis is based primarily on clinical manifestations, such as thecolor around the lesion or wound, swelling, <strong>to</strong>xemia, <strong>and</strong> muscle tissue destruction.The presence of gas is not always indicative of clostridial infection. A smear of exudatefrom the wound or a gram-stained muscle tissue sample may be helpful in diagnosisif numerous large gram-positive bacilli are found. The culture of anaerobicbacilli from human cases is generally of little value because of the time required <strong>and</strong>the urgency of diagnosis. Moreover, isolation of a potentially pathogenic anaerobefrom a wound may only indicate contamination <strong>and</strong> not necessarily active infection(penetration <strong>and</strong> multiplication in the human or animal organism). In animals, culturecan be important in distinguishing infection caused by C. chauvoei (symp<strong>to</strong>maticanthrax, blackleg, or emphysema<strong>to</strong>us gangrene) from infections caused by C.septicum. The latter bacterium rapidly invades the animal’s body after death; thus,the material used for examination should be taken before or very shortly after death.The fluorescent antibody technique permits identification of the pathogenicclostridia in a few hours <strong>and</strong> can be very useful in diagnosis.Control: Prevention of the infection consists of prompt treatment of wounds <strong>and</strong>removal of foreign bodies <strong>and</strong> necrotic tissue. Special care must be taken <strong>to</strong> ensurethat <strong>to</strong>urniquets, b<strong>and</strong>ages, <strong>and</strong> casts do not interfere with circulation <strong>and</strong> thus createconditions favorable <strong>to</strong> the multiplication of anaerobic bacteria by reducing localoxidation-reduction potential.Combined vaccines of C. chauvoei <strong>and</strong> C. septicum are used for active immunizationof calves <strong>and</strong> lambs. Vaccination with bacterins or alpha <strong>to</strong>xoid must be carriedout prior <strong>to</strong> castration, tail docking, shearing, or removal of horns. Calves canbe vaccinated at 2 months of age.BibliographyBartlett, J.G. Gas gangrene (other clostridium-associated <strong>diseases</strong>). In: M<strong>and</strong>ell, G.L., R.G.Douglas, Jr., J.E. Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. NewYork: Churchill Livings<strong>to</strong>ne, Inc.; 1990.Bruner, D.W., J.H. Gillespie. Hagan’s Infectious Diseases of Domestic Animals. 6th ed.Ithaca: Coms<strong>to</strong>ck; 1973.Finegold, S.M. Anaerobic Bacteria in Human Disease. New York: Academic Press; 1977.Joklik, W.K., D.T. Smith. Zinsser’s Microbiology. 15th ed. New York: Meredith; 1972.MacLennan, J.D. The his<strong>to</strong><strong>to</strong>xic clostridial infections of man. Bact Rev 26:177–274, 1962.Prévot, A.R., A. Turpin, P. Kaiser. Les bactéries anaerobies. Paris: Dunod; 1967.Rose, H.M. Disease caused by clostridia. In: Wyngaarden, J.B., L.H. Smith, Jr., eds. CecilTextbook of Medicine. 16th ed. Philadelphia: Saunders; 1982.Rosen, M.M. Clostridial infections <strong>and</strong> in<strong>to</strong>xications. In: Hubbert, W.T., W.F. McCulloch,P.R. Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield:Thomas; 1975.Smith, D.L.S. Clostridial <strong>diseases</strong> of animals. Adv Vet Sci 3:465–524, 1957.Smith, L.D., L.V. Holderman. The Pathogenic Anaerobic Bacteria. Springfield:Thomas; 1968.


90 BACTERIOSESCOLIBACILLOSISICD-10 A04.0 enteropathogenic Escherichia coli infection;A04.1 entero<strong>to</strong>xigenic Escherichia coli infection;A04.2 enteroinvasive Escherichia coli infection;A04.3 enterohemorrhagic Escherichia coli infectionSynonyms: Colibacteriosis, coli<strong>to</strong>xemia, enteropathogenic diarrhea.Etiology <strong>and</strong> Physiopathogenesis: Escherichia coli belongs <strong>to</strong> the familyEnterobacteriaceae. E. coli is a normal component of the flora in the large intestineof warm-blooded animals, including man. It is a gram-negative, motile or nonmotile,facultatively anaerobic bacillus.It is classified in<strong>to</strong> different serotypes according <strong>to</strong> the scheme originally developedby Kauffmann, which is based primarily on the somatic O antigens (polysaccharide<strong>and</strong> thermostable) that differentiate E. coli in<strong>to</strong> more than 170 serogroups.The flagellar H antigen, which is thermolabile <strong>and</strong> proteinic, distinguishes theserotypes (56 <strong>to</strong> date) of each serogroup. The K (capsular) <strong>and</strong> F (fimbrial) antigensare also important (Doyle <strong>and</strong> Padhye, 1989). The pathogenic strains, which causeenteric disease, are grouped in<strong>to</strong> six categories: (a) enterohemorrhagic (EHEC), (b)entero<strong>to</strong>xigenic (ETEC), (c) enteroinvasive (EIEC), (d) enteropathogenic (EPEC),(e) enteroaggregative (EAggEC), <strong>and</strong> (f) diffuse-adherent (DAEC). The last two categoriesare not yet well defined, <strong>and</strong> the last category is not dealt with here. Thesecategories differ in their pathogenesis <strong>and</strong> virulence properties, <strong>and</strong> each comprisesa distinct group of O:H serotypes. Their clinical symp<strong>to</strong>ms <strong>and</strong> epidemiological patternsmay also differ (Chin, 2000).In terms of the <strong>zoonoses</strong>, the most important category is the enterohemorrhagic,which is also the most severe.a) Enterohemorrhagic E. coli (EHEC). The principal etiologic agent of thiscolibacillosis is E. coli O157:H7. Since it was first recognized in 1983 (Riley et al.,1983), this category has been a public health problem in Europe <strong>and</strong> the US, whichbecame more serious with an outbreak that occurred in the latter betweenNovember 15, 1992 <strong>and</strong> February 28, 1993. In Washing<strong>to</strong>n State <strong>and</strong> other westernUS states, 470 people fell ill <strong>and</strong> four died—three in Washing<strong>to</strong>n <strong>and</strong> one in SanDiego, California (Spencer, 1993; Dorn, 1993). Griffin <strong>and</strong> Tauxe (1991) concludethat O157:H7 is an emerging <strong>and</strong> new pathogen, because they feel that such a distinctiveillness—which often has serious consequences (hemolytic uremic syndrome)—wouldhave attracted attention in any period. Later, O26:H11, O45:H2,<strong>and</strong> three nonmotile E. coli—O4, O111, <strong>and</strong> O145—were added <strong>to</strong> this serotype.This group is characterized by a 60-megadal<strong>to</strong>n virulence plasmid <strong>and</strong> by its secretionof Shiga-like <strong>to</strong>xins or vero<strong>to</strong>xins. The Shiga-like <strong>to</strong>xin was thus namedbecause it is similar in structure <strong>and</strong> activity <strong>to</strong> the <strong>to</strong>xin produced by Shigella disenteriaetype 1, <strong>and</strong> is neutralized by the Shiga <strong>to</strong>xin antiserum. There are actuallytwo <strong>to</strong>xins, Shiga-like <strong>to</strong>xin I <strong>and</strong> Shiga-like <strong>to</strong>xin II (or vero<strong>to</strong>xins I <strong>and</strong> II). Bothare cy<strong>to</strong><strong>to</strong>xic (lethal <strong>to</strong> Vero <strong>and</strong> HeLa cells), cause fluid accumulation in rabbit ligatedileal loops, <strong>and</strong> paralysis <strong>and</strong> death in mice <strong>and</strong> rabbits (O’Brien <strong>and</strong> Holmes,1987). Vero<strong>to</strong>xin II produces hemorrhagic colitis in adult rabbits. The two <strong>to</strong>xins areantigenically different.


COLIBACILLOSIS 91Geographic Distribution <strong>and</strong> Occurrence in Man: Worldwide. SerotypeO157:H7 has been isolated in outbreaks in Canada, Great Britain, <strong>and</strong> the UnitedStates. It has also been isolated in Argentina, Australia, Belgium, the formerCzechoslovakia, China, Germany, Holl<strong>and</strong>, Irel<strong>and</strong>, Italy, Japan, <strong>and</strong> South Africa(Griffin <strong>and</strong> Tauxe, 1991). These isolates were obtained from fecal samples takenfrom sporadic cases of hemorrhagic diarrhea submitted <strong>to</strong> public health or hospitallabora<strong>to</strong>ries for examination.From 1982 <strong>to</strong> 1992, 17 outbreaks occurred in the US; the smallest affected 10people <strong>and</strong> the largest 243. In November 1992, an outbreak occurred among peoplewho had eaten undercooked hamburgers at a fast food restaurant chain. Thesame E. coli serotype was isolated from the ground beef found in these restaurants(CDC, 1993). Seventeen more outbreaks occurred in 1993. Case-reporting is nowcompulsory in 18 US states. It is estimated that there are 8 cases each year per100,000 inhabitants in Washing<strong>to</strong>n State (approximately the same incidence as forsalmonellosis).During the same period (1982–1992), there were three outbreaks in Canada <strong>and</strong>two in Great Britain (Griffin <strong>and</strong> Tauxe, 1991).Occurrence in Animals: Based on outbreaks in the US, studies were conducted<strong>to</strong> evaluate the infection rate in cattle. The agent was isolated from only 25 sucklingcalves of the approximately 7,000 examined in 28 states. This study indicated thatthe agent is widely distributed in the US, but that the rate of animals harboring thisserotype is low. The prevalence of infected herds is estimated at approximately5%. In Washing<strong>to</strong>n State, between 5% <strong>and</strong> 10% of herds harbor E. coli O157:H7(Spencer, 1993). This serotype was also isolated from cattle in Argentina, Canada,Egypt, Germany, Great Britain, <strong>and</strong> Spain. In Argentina <strong>and</strong> Spain, there wasan association between serotype O157:H7 <strong>and</strong> a diarrheal disease in cattle, whereasin the other countries the isolates were produced from apparently normal cattle(Dorn, 1993).The Disease in Man: The incubation period is from two <strong>to</strong> nine days. The appearanceof the disease ranges from a slight case of diarrhea <strong>to</strong> severe hemorrhagic colitis,with strong abdominal pains <strong>and</strong> little or no fever. At the outset, diarrhea iswatery but later becomes hemorrhagic, either with traces of blood or highly hemorrhagics<strong>to</strong>ols. Diarrhea lasts an average of four days <strong>and</strong> about 50% of patients experiencevomiting. Hemorrhagic diarrhea was present in more than 95% of a largenumber of sporadic cases recorded. In some outbreaks in nursing homes, wherestricter surveillance was possible, it was shown that between 56% <strong>and</strong> 75% ofaffected patients had hemorrhagic s<strong>to</strong>ols <strong>and</strong> the rest had diarrhea without blood;asymp<strong>to</strong>matic infections were also confirmed (Griffin <strong>and</strong> Tauxe, 1991). E. coliO157:H7 infection is feared primarily because of its complications. One of these ishemolytic uremic syndrome, which is the principal cause of acute renal deficiencyin children <strong>and</strong> frequently requires dialysis <strong>and</strong> transfusions. Another complicationis thrombotic thrombocy<strong>to</strong>penic purpura, which is characterized by thrombocy<strong>to</strong>penia,hemolytic anemia, azotemia, fever, thrombosis in the terminal arterioles <strong>and</strong>capillaries, <strong>and</strong> neurological symp<strong>to</strong>ms that dominate the clinical picture.Depending on the population, cases involving hemolytic uremic syndrome probablyrepresent between 2% <strong>and</strong> 7% of the <strong>to</strong>tal number of cases due <strong>to</strong> E. coli O157:H7(Griffin <strong>and</strong> Tauxe, 1991).


92 BACTERIOSESAlthough E. coli O157:H7 is susceptible <strong>to</strong> many <strong>common</strong>ly used antibiotics, theyshould not be used as a preventive measure. During an outbreak in a nursing home,antibiotics were considered a risk fac<strong>to</strong>r for contracting infection (Carter et al.,1987). It is believed that antibiotics may increase the risk of infection <strong>and</strong> complications,probably by stimulating the production of <strong>to</strong>xin <strong>and</strong> altering the normalintestinal flora, thus allowing greater growth of serotype O157:H7. There is also arisk of producing resistant strains (Dorn, 1993).Source of Infection <strong>and</strong> Mode of Transmission: Of nine outbreaks in the US,six were caused by undercooked ground beef <strong>and</strong> three by roast beef. An outbreakin Canada was caused by raw milk. These facts point <strong>to</strong> cattle as the reservoir of theEHEC agent. Other foods, such as cold s<strong>and</strong>wiches <strong>and</strong> uncooked pota<strong>to</strong>es, werealso investigated; calf feces was the suspected contaminant in the pota<strong>to</strong>es. A laterstudy indicated that undercooked meat (especially from calves <strong>and</strong> heifers) was thesource of infection in more than 75% of the outbreaks. Another outbreak tha<strong>to</strong>ccurred in 1989 in Cabool, Missouri (USA) <strong>and</strong> affected 243 people (one of every12 people in the <strong>to</strong>wn) was caused by city-supplied water. The water may have beencontaminated by deer feces. Human-<strong>to</strong>-human transmission also occurs, as secondarycases, through the fecal-oral route. A baby-sitter contracted the infection whilecaring for a sick child. Secondary cases have also occurred in day-care centers(Dorn, 1993).Diagnosis: Sorbi<strong>to</strong>l-MacConkey (SMAC) agar is recommended for isolation ofE. coli O157:H7 from fecal samples. Various enzyme immunoassay techniques canbe used <strong>to</strong> detect Shiga-like <strong>to</strong>xins in fecal matter or cultures. Isolation becomes difficultbeyond one week after the onset of symp<strong>to</strong>ms.Control <strong>and</strong> Prevention: Ground beef should be cooked until it is no longerpink. Meat from cattle, like that of other mammalian <strong>and</strong> avian species, can be contaminatedby feces during slaughter <strong>and</strong> processing. Thus, all precautions should betaken <strong>to</strong> minimize this risk, <strong>and</strong> foods of animal origin should be well cooked beforethey are eaten. Personal hygiene, particularly h<strong>and</strong>washing after relieving oneself, isalso important (Doyle <strong>and</strong> Padhye, 1989).b) Entero<strong>to</strong>xigenic E. coli (ETEC). Entero<strong>to</strong>xigenic E. coli has been the categorymost intensely studied in recent years. Research has not only added knowledgeabout the physiopathogenic action mechanisms of these bacteria, but has also providedmeans <strong>to</strong> prevent diarrheal disease in several animal species. Entero<strong>to</strong>xigenicstrains synthesize various types of <strong>to</strong>xins—a heat-labile (LT) <strong>to</strong>xin that is immunologicallyrelated <strong>to</strong> the cholera <strong>to</strong>xin, a heat-stable (ST) <strong>to</strong>xin that is not antigenic,or both (LT/ST). The <strong>to</strong>xins are plasmid-dependent <strong>and</strong> may be transferable fromone ETEC strain <strong>to</strong> other strains that lack them.Entero<strong>to</strong>xigenic strains are distributed heterogeneously among the different O:Hserotypes. ETEC strains make use of fimbriae or pili (nonflagellar, proteinic, filamen<strong>to</strong>usappendices) <strong>to</strong> adhere <strong>to</strong> the mucosa of the small intestine, multiply, <strong>and</strong>produce one or more <strong>to</strong>xins. These pili interact with epithelial cells, are very importantvirulence elements, <strong>and</strong> are called colonization fac<strong>to</strong>rs. Since the <strong>to</strong>xins areplasmid-dependent, the antigenic characteristics of the pili differ in different animalspecies. In man, there are seven colonization fac<strong>to</strong>rs: CFA-1 <strong>and</strong> CS1 through CS6(<strong>WHO</strong>, 1991). In calves <strong>and</strong> lambs, the implicated pili are primarily F5 (formerly


COLIBACILLOSIS 93K99). Although F4 (K88) <strong>and</strong> 987P have also been isolated, they are not believed <strong>to</strong>play a role in ETEC virulence in these animals. The pili associated with entero<strong>to</strong>xigeniccolibacillosis in suckling pigs are F4 (K88), F5 (K99), F41, <strong>and</strong> 987P.In the developing countries, the entero<strong>to</strong>xigenic E. coli group primarily affectschildren under 2 or 3 years of age. In unhygienic homes, children may frequently sufferfrom ETEC. The incidence of the disease declines after the age of 4 <strong>and</strong> remainslow. In addition, ETEC is the most <strong>common</strong> cause of “traveller’s diarrhea” in adultswho visit endemic countries. This epidemiological characteristic suggests that thepopulation in endemic countries acquires immunity, while in industrialized countriesthe population is little exposed <strong>to</strong> these agents <strong>and</strong> does not acquire immunity.The disease in man produces symp<strong>to</strong>ms that closely resemble those caused byVibrio cholerae. After an incubation period of 12 <strong>to</strong> 72 hours, there is profuse,watery diarrhea; abdominal colic; vomiting; acidosis; <strong>and</strong> dehydration. The feces donot contain mucus or blood <strong>and</strong> there may be fever. The duration of the illness isshort <strong>and</strong> the symp<strong>to</strong>ms generally disappear in two <strong>to</strong> five days.ETEC can be diagnosed in man by demonstrating the presence of entero<strong>to</strong>xin TL,TS, or both through an enzyme immunoassay. DNA probes can also be used <strong>to</strong> identifythe genes in the bacteria that encode the <strong>to</strong>xins.Man is the main reservoir <strong>and</strong> source of infection is the feces of patients <strong>and</strong> carriers.The route of transmission is fecal-oral. The vehicle of infection may be food<strong>and</strong> water contaminated by human feces.ETEC is the cause of some outbreaks that affected many people in the developedcountries. Some occurred in children’s hospitals in Great Britain <strong>and</strong> the US,although the source of infection was not definitively determined. There have beenoutbreaks among adults that affected hundreds of people <strong>and</strong> were attributed <strong>to</strong> specificfoods <strong>and</strong> contaminated water. One of the largest outbreaks, affecting morethan 2,000 people, occurred in 1975 in a national park in Oregon (USA). Other outbreakswere due <strong>to</strong> imported Brie cheese that caused enterocolitis in several USstates as well as in Denmark, the Netherl<strong>and</strong>s, <strong>and</strong> Sweden. A large outbreak affecting400 people occurred among diners at a restaurant in Wisconsin (USA). In thiscase, the source of infection was believed <strong>to</strong> be an employee who had diarrhea twoweeks prior <strong>to</strong> the outbreak. A passenger on a cruise ship suffered two episodes ofgastroenteritis caused by ETEC, one of them due <strong>to</strong> the ship’s contaminated water(Doyle <strong>and</strong> Padhye, 1989).c) Enteroinvasive E. coli (EIEC). This category represents a small group of E.coli. Many components are nonmotile (lacking the H antigen) <strong>and</strong> they are slow <strong>to</strong>ferment lac<strong>to</strong>se or are nonlac<strong>to</strong>se fermenting. The disease they cause is very similar<strong>to</strong> bacillary dysentery caused by Shigella. Their somatic antigens may cross-reactwith those of Shigella. Enteroinvasive E. coli can invade <strong>and</strong> multiply in the cells ofthe intestinal mucosa, especially in the colon.EIEC colitis begins with strong abdominal pains, fever, malaise, myalgia,headache, <strong>and</strong> watery feces containing mucus <strong>and</strong> blood. The incubation period isfrom 10 <strong>to</strong> 18 hours. If diarrhea is severe, the patient can be treated with ampicillin.The reservoir seems <strong>to</strong> be man <strong>and</strong> the source of infection contaminated water orfood. However, the source of infection is not always definitively identified.EIEC is endemic in the developing countries <strong>and</strong> accounts for 1% <strong>to</strong> 5% of allpatients with diarrhea who see a doc<strong>to</strong>r (Benenson, 1990). Studies conducted with


94 BACTERIOSESvolunteers indicate that a very high bacterial load is needed <strong>to</strong> reproduce the disease.Some outbreaks due <strong>to</strong> contaminated water <strong>and</strong> food have occurred in the developedcountries.EIEC can be suspected when a large number of leukocytes is found in a preparationmade from fecal mucus. The guinea pig-kera<strong>to</strong>conjunctivitis test (Sereny test)has diagnostic value. This test uses enteroinvasive E. coli cultures <strong>to</strong> demonstrate thecapacity <strong>to</strong> invade epithelial cells. An enzyme immunoassay has been developed <strong>to</strong>detect a polypeptide in the surface membrane of the bacteria that determines virulence(invasive capacity).d) Enteropathogenic E. coli (EPEC). The etiologic agents of the enteropathogenicdisease belong <strong>to</strong> 15 O serogroups of E. coli. The disease occurs primarily innursing babies under 1 year, in whom it can cause a high mortality rate.The disease is characterized by watery diarrhea containing mucus but no visibleblood; fever; <strong>and</strong> dehydration. The incubation period is short.The disease occurs primarily in developing countries <strong>and</strong> has practically disappearedin Europe <strong>and</strong> the US. It occurs mostly in the warm seasons (summer diarrhea)<strong>and</strong> the sources of infection are formula milk <strong>and</strong> weaning foods that becomecontaminated due <strong>to</strong> poor cleaning of bottles <strong>and</strong> nipples, or deficient hygiene on themother’s part. Children in poor socioeconomic groups are frequently exposed <strong>to</strong>EPEC <strong>and</strong> generally acquire immunity after the first year of life. In epidemic diarrheain newborns in nurseries, airborne transmission is possible through contaminateddust. Some outbreaks have also been described in adults.E. coli isolated from feces must be serotyped. Once the EPEC serotype has beendetermined, a DNA probe should be used <strong>to</strong> try <strong>to</strong> identify the EPEC adherence fac<strong>to</strong>r(EAF), which is plasmid-dependent. EPEC strains also show localized adherence<strong>to</strong> HEp-2 cells.In epidemics, hospitals <strong>and</strong> nurseries should have a separate room for sick babies.Treatment consists primarily of electrolyte replacement with oral saline solutions, orwith intravenous solutions, if necessary. In most cases, no other treatment is needed.In serious cases, the child can be given oral cotrimoxazole, which reduces the intensity<strong>and</strong> duration of the diarrhea. Feeding, including breast-feeding, should continue(Benenson, 1990).e) Enteroaggregative E. coli (EAggEC). This name is given <strong>to</strong> a group of E. colithat has an aggregative adherence pattern in an HEp-2 assay rather than a localized(as in EPEC) or diffuse one. This category is provisional until it is better defined. Astudy was done on 42 cultures—40 from children with diarrhea in Santiago (Chile),1 from Peru, <strong>and</strong> 1 from a North American student who had visited Mexico. Allthese strains tested negative for enterohemorrhagic, entero<strong>to</strong>xigenic, enteropathogenic,<strong>and</strong> enteroinvasive E. coli with DNA probes. They also failed <strong>to</strong> fit in one ofthese categories on the basis of serotyping. This group causes characteristic lesionsin rabbit ligated ileal loops <strong>and</strong> mice (Vial et al., 1988; Levine et al., 1988).EAggEC causes persistent diarrhea in nursing babies. The incubation period isestimated at one <strong>to</strong> two days (Benenson, 1990).The Disease in Animals: In addition <strong>to</strong> sporadic cases of mastitis, urogenitalinfections, abortions, <strong>and</strong> other pathological processes, E. coli is responsible for severalimportant <strong>diseases</strong>.


COLIBACILLOSIS 95CATTLE: Calf diarrhea (white scours) is an acute disease that causes high mortalityin calves less than 10 days old. It manifests as serious diarrhea, with whitish feces <strong>and</strong>rapid dehydration. It may last from a few hours <strong>to</strong> a few days. Colostrum-deprivedcalves are almost always victims of this disease. Colostrum, with its high IgM content,is essential in preventing diarrhea in calves. In the first 24 <strong>to</strong> 36 hours of life, the intestinalmembrane is permeable <strong>to</strong> immunoglobulins, which pass quickly in<strong>to</strong> the bloodstream<strong>and</strong> protect the animal against environmental microorganisms.Entero<strong>to</strong>xigenic strains that cause diarrhea in newborn calves are different fromhuman strains. They generally produce a heat-stable <strong>to</strong>xin <strong>and</strong> the pili antigen isalmost always type F5 (K99).The septicemic form of colibacillosis in colostrum-deprived calves includes diarrheaas well as signs of generalized infection. Animals that survive longer usuallysuffer from arthritis <strong>and</strong> meningitis (Gillespie <strong>and</strong> Timoney, 1981).Mastitis due <strong>to</strong> E. coli appears particularly in older cows with dilated milk ducts. Inmilk without leukocytes, coliforms multiply rapidly, causing an inflamma<strong>to</strong>ry reactionthat destroys the bacteria <strong>and</strong> releases a large quantity of endo<strong>to</strong>xins. This producesacute mastitis, with fever, anorexia, cessation of milk production, <strong>and</strong> weight loss. Inthe next lactation period, the mammary gl<strong>and</strong>s return <strong>to</strong> normal function.SHEEP: A disease with white diarrhea, similar <strong>to</strong> that in calves, has been reportedin lambs in several countries. In South Africa, colipathogens were indicated as thecause of a septicemic illness in lambs, with neurological symp<strong>to</strong>ms, ascites, <strong>and</strong>hydropericarditis, but without major gastrointestinal disorders.HORSES: A long-term study of horse fetuses <strong>and</strong> newborn colts found that close <strong>to</strong>1% of abortions <strong>and</strong> 5% of newborn deaths were due <strong>to</strong> E. coli.SWINE: Neonatal enteritis in suckling pigs, caused by E. coli,begins 12 hours afterbirth with profuse, watery diarrhea <strong>and</strong> may end with fatal dehydration. Mortality isparticularly high in suckling pigs from sows giving birth for the first time. About50% of isolated strains are <strong>to</strong>xicogenic <strong>and</strong> some produce both thermostable (ST)<strong>and</strong> thermolabile (LT) <strong>to</strong>xins (Gillespie <strong>and</strong> Timoney, 1981). In newborn piglets, thecolonization fac<strong>to</strong>rs are F4 (K88), F5 (K99), F41, <strong>and</strong> 987P <strong>and</strong> there is probablyone other fac<strong>to</strong>r. Diarrhea in weaned piglets is caused by hemolytic strains of ETECthat have the F4 (K88) colonization fac<strong>to</strong>r, but there are also some strains thatexpress no known fac<strong>to</strong>r (Casey et al., 1992). Diarrhea begins shortly after weaning<strong>and</strong> is a very <strong>common</strong> complication. The animals also suffer from anorexia <strong>and</strong>depression. Mortality is lower than in newborn suckling pigs <strong>and</strong> the pathogenesismay be similar.Edema in suckling pigs is an acute disease that generally attacks between 6 <strong>and</strong> 14weeks of age. It is becoming increasingly important in swine-producing areas. It ischaracterized by sudden onset, uncoordinated movement, <strong>and</strong> edema of the eyelids,the cardiac region of the s<strong>to</strong>mach, <strong>and</strong> occasionally other parts of the body. Bodytemperature is usually normal. Neurological symp<strong>to</strong>ms may be preceded by diarrhea(Nielsen, 1986). The disease usually occurs in winter. Morbidity ranges from 10% <strong>to</strong>35% <strong>and</strong> mortality from 20% <strong>to</strong> 100%. The disease seems <strong>to</strong> be triggered by stressdue <strong>to</strong> weaning, changes in diet, <strong>and</strong> vaccination against hog cholera. The diseasemechanism could be an intestinal <strong>to</strong>xemia caused by specific strains of E. coli. A variant<strong>to</strong>xin similar <strong>to</strong> Shiga-like <strong>to</strong>xin II (see enterohemorrhagic E. coli) was identified


96 BACTERIOSESas the principal fac<strong>to</strong>r in the edema. This variant is <strong>to</strong>xic for Vero cells, but not HeLacells (Dobrescu, 1983; Marques et al., 1987; Kausche et al., 1992).FOWL: Pathogenic serotypes of E. coli have been isolated in septicemic <strong>diseases</strong>of fowl, as well as in cases of salpingitis <strong>and</strong> pericarditis. Contamination of eggs byfeces or through ovarian infection is the source of colibacillosis in newborn chicks.Colibacillosis in adult chickens <strong>and</strong> turkeys primarily affects the lungs, though itmay also invade the circula<strong>to</strong>ry system <strong>and</strong> cause septicemia <strong>and</strong> death (Timoney etal., 1988). Another avian disease, “swollen head” syndrome, has also beendescribed. It is characterized by swelling of the orbital sinuses, <strong>to</strong>rticollis,opistho<strong>to</strong>nos, <strong>and</strong> a lack of coordination. The illness lasts two <strong>to</strong> three weeks, <strong>and</strong>mortality is between 3% <strong>and</strong> 4% (O’Brien, 1985). Its etiology is uncertain. Viruses,E. coli, <strong>and</strong> some other bacteria have been isolated. The viral infection (paramyxovirus,coronavirus, pneumovirus) is thought <strong>to</strong> cause acute rhinitis <strong>and</strong> prepare theway for E. coli <strong>to</strong> invade subcutaneous facial tissue. It was possible <strong>to</strong> reproduce thedisease with some strains of E. coli; in contrast, the disease could not be reproducedwith the viruses (White et al., 1990; Pages Mante <strong>and</strong> Costa Quintana, 1987). A colibacillaryetiology has also been attributed <strong>to</strong> Hjarre’s disease (coligranuloma),which causes granuloma<strong>to</strong>us lesions in the liver, cecum, spleen, bone marrow, <strong>and</strong>lungs of adult fowl. The lesions resemble those of tuberculosis <strong>and</strong> mucoid strainsof E. coli have been isolated from them. The disease can be reproduced in labora<strong>to</strong>ryanimals <strong>and</strong> chickens by parenteral inoculation but not by oral administration.Source of Infection <strong>and</strong> Mode of Transmission: Man is the reservoir for all categoriesexcept enterohemorrhagic E. coli (EHEC), for which there are strong indicationsthat the reservoir is cattle.Cattle <strong>and</strong> swine may occasionally harbor strains of entero<strong>to</strong>xigenic E. coli(ETEC) in their intestines, as Doyle <strong>and</strong> Padhye (1989) point out. In Bangladesh,three ETEC cultures were isolated from healthy calves <strong>and</strong> cows; these were of thesame serotype <strong>and</strong> <strong>to</strong>xin variety as those taken from patients with diarrhea who hadbeen in contact with the animals (Black et al., 1981). In the Philippines, an ETECserotype (O78:H12, LT + ST + ) was isolated from a rectal swab from a pig; this serotypewas considered <strong>to</strong> be the agent of human diarrhea in many countries (Echeverría etal., 1978, cited in Doyle <strong>and</strong> Padhye, 1989). However, volunteers were fed an ETECstrain isolated from a pig, but none of them had diarrhea (Du Pont et al., 1971, citedin Doyle <strong>and</strong> Padhye, 1989). The source of infection is the feces of infected persons(primarily sick persons, secondarily carriers) <strong>and</strong> objects contaminated by them. Themost <strong>common</strong> mode of transmission is the oral-fecal route. Contaminated foods,including those from animals (meat, milk, cheeses), are a <strong>common</strong> vehicle in variouscategories of human colibacillosis. In EHEC, beef is considered the principal sourceof human infection. In the case of epidemic diarrhea in newborn infants in nurseries,airborne transmission by contaminated dust is possible.In animals, the source of infection <strong>and</strong> mode of transmission follow the same patternsas in human infection. Animals with diarrhea constitute the main source ofinfection.Diagnosis: Diagnosis in man is based on isolation of the etiologic agent <strong>and</strong> on teststhat can identify it as enterohemorrhagic, entero<strong>to</strong>xigenic, enteroinvasive, or enteroaggregative(see each separate category for the most suitable diagnostic method).


COLIBACILLOSIS 97In the case of diarrhea in newborn cattle, sheep, <strong>and</strong> swine, fresh feces or theintestinal contents of a recently dead or slaughtered animal can be cultured.The immunofluorescence test is very useful for detecting colonization fac<strong>to</strong>rs;sections of the ileal loop of a recently dead animal are stained with conjugate for thispurpose (Timoney et al., 1988).Control: For man, control measures include: (a) personal cleanliness <strong>and</strong>hygienic practices, sanitary waste removal, <strong>and</strong> environmental sanitation; (b) provisionof maternal <strong>and</strong> child hygiene services; (c) protection of food products, pasteurizationof milk, <strong>and</strong> compulsory veterinary inspection of meat; <strong>and</strong> (d) specialpreventive measures in hospital nursery wards. These measures should include keepinghealthy newborns separate from sick nursing infants or older children. Nurseswho tend the nurseries should not have contact with other wards, <strong>and</strong> those in chargeof feeding bottles should not change diapers. Special precautions should be taken inthe laundry.To prevent colibacillosis in animals, the <strong>common</strong>ly accepted rules of herd managementshould be followed. For calves, colostrum is important for the preventionof white scours, <strong>and</strong> for pigs, all unnecessary stress should be avoided during weaningin order <strong>to</strong> prevent edema.In recent years, investigations of the fac<strong>to</strong>rs that permit entero<strong>to</strong>xigenic E. colistrains <strong>to</strong> colonize the small intestine have opened up new horizons in colibacillosisprevention in animals. Vaccines for cattle <strong>and</strong> swine have been developed based onfimbria (pili) antigens. These antigens inhibit E. coli from adhering <strong>to</strong> the mucosa ofthe small intestine. To this end, gestating cows <strong>and</strong> sows are vaccinated with vaccinesbased on F5 (K99) <strong>and</strong> F4 (K88) antigens, respectively. Newborns acquire passiveimmunity via colostrum <strong>and</strong> milk, which contain antibodies against these fac<strong>to</strong>rs. Inthe same way, good results have been obtained in protecting newborn lambs by vaccinatingewes with F5 (K99). In addition, studies (Rutter et al., 1976; Myers, 1978;Nagy, 1980) are being carried out with oral vaccines for humans using <strong>to</strong>xicogenicE. coli <strong>to</strong>xoids of both thermolabile <strong>and</strong> thermostable <strong>to</strong>xins as well as antiadherencefac<strong>to</strong>rs (purified fimbriae). Genetic engineering is another approach being used <strong>to</strong>obtain vaccines with attenuated E. coli virulence (Levine <strong>and</strong> Lanata, 1983).BibliographyBenenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Biester, H.E., L.H. Schwarte, eds. Diseases of Poultry. 4th ed. Ames: Iowa State UniversityPress; 1959.Binsztein, N. Estudio de la diarrea. Fac<strong>to</strong>res de virulencia <strong>and</strong> mecanismos fisiopa<strong>to</strong>génicos.Bacteriol Clin Argent 1:138–142, 1982.Black et al., 1981. Cited in: Doyle, M.P., V.V. Padhye. Escherichia coli. In:Doyle, M.P., ed.Foodborne Bacterial Pathogens. New York: Marcel Dekker; 1989.Carter, A.O., A.A. Borczyk, J.A. Carlson, et al. A severe outbreak of Escherichiacoli O157:H7-associated hemorrhagic colitis in a nursing home. N Engl J Med317:1496–1500, 1987.Casey, T.A., B. Nagy, H.W. Moon. Pathogenicity of porcine entero<strong>to</strong>xigenic Escherichiacoli that do not express K88, K99, F41, or 987P adhesins. Am J Vet Res 53:1488–1492, 1992.


98 BACTERIOSESChin, J., ed. Control of Communicable Diseases Manual. 17th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 2000.Dobrescu, L. New biological effect of edema disease principle (Escherichia coli neuro<strong>to</strong>xin)<strong>and</strong> its use as an in vitro assay for this <strong>to</strong>xin. Am J Vet Res 44:31–34, 1983.Dorn, C.R. Review of foodborne outbreak of Escherichia coli O157:H7 infection in thewestern United States. J Am Vet Med Assoc 203:1583–1587, 1993.Doyle, M.P., V.V. Padhye. Escherichia coli. In: Doyle, M.P., ed. Foodborne BacterialPathogens. New York: Marcel Dekker; 1989.Du Pont et al., 1971. Cited in: Doyle, M.P., V.V. Padhye. Escherichia coli. In: Doyle, M.P.,ed. Foodborne Bacterial Pathogens. New York: Marcel Dekker; 1989.Echeverría et al., 1978. Cited in: Doyle, M.P., V.V. Padhye. Escherichia coli. In: Doyle,M.P., ed. Foodborne Bacterial Pathogens. New York: Marcel Dekker; 1989.Edwards, P.R., W.H. Ewing. Identification of Enterobacteriaceae. 3rd ed. Minneapolis:Burgess; 1972.Ellens, D.J., P.W. de Leeuw, H. Rozemond. Detection of the K99 antigen of Escherichiacoli in calf feces by enzyme-linked immunosorbent assay (ELISA). Tijdschr Dieregeneedskd104:169–175, 1979.Fraser, C.M., J.A. Bergeron, A. Mays, S.E. Aiello, eds. The Merck Veterinary Manual. 7thed. Rahway: Merck; 1991.Gay, C.C. Escherichia coli <strong>and</strong> neonatal disease of calves. Bact Rev 29:75–101, 1965.Gillespie, J.H., J.F. Timoney. Hagan <strong>and</strong> Bruner’s Infectious Diseases of DomesticAnimals. 7th ed. Ithaca: Coms<strong>to</strong>ck; 1981.Griffin, P.M., R.V. Tauxe. The epidemiology of infections caused by Escherichia coliO157:H7, other enterohemorrhagic E. coli, <strong>and</strong> the associated hemolytic uremic syndrome.Epidemiol Rev 13:60–98, 1991.Kausche, F.M., E.A. Dean, L.H. Arp, et al. An experimental model for subclinical edemadisease (Escherichia coli entero<strong>to</strong>xemia) manifest as vascular necrosis in pigs. Am J Vet Res53:281–287, 1992.Levine, M.M., C. Lanata. Progresos en vacunas contra diarrea bacteriana. Adel MicrobiolEnf Infec 2:67–118, 1983.Levine, M.M, V. Prado, R. Robins-Browne, et al. Use of DNA probes <strong>and</strong> HEp-2 celladherence assay <strong>to</strong> detect diarrheagenic Escherichia coli. J Infect Dis 158:224–228, 1988.Marques, L.R.M., J.S.M. Peiris, S.J. Cryz, et al. Escherichia coli strains isolated from pigsproduce a variant Shiga-like <strong>to</strong>xin II. FEMS Microbiol Lett 44:33–38, 1987.Merson, M.H., R.H. Yolken, R.B. Sack, J.L. Froehlich, H.B. Greenberg, I. Huq, et al.Detection of Escherichia coli entero<strong>to</strong>xins in s<strong>to</strong>ols. Infect Immun 29:108–113, 1980.Mills, K.W., K.L. Tietze, R.M. Phillips. Use of enzyme-linked immunosorbent assay fordetection of K88 pili in fecal specimens from swine. Am J Vet Res 44:2188–2189, 1983.Myers, L.L. Enteric colibacillosis in calves: immunogenicity <strong>and</strong> antigenicity ofEscherichia coli isolated from calves with diarrhea. Infect Immun 13:1117–1119, 1978.Nagy, B. Vaccination of cows with a K99 extract <strong>to</strong> protect newborn calves against experimentalentero<strong>to</strong>xic colibacillosis. Infect Immun 27:21–24, 1980.Nielsen, N.O. Edema disease. In: Leman, A.D., B. Straw, R.D. Glock, et al., eds. Diseasesof Swine. 6th ed. Ames: Iowa State University Press; 1986.O’Brien, A.D., R.K. Holmes. Shiga <strong>and</strong> Shiga-like <strong>to</strong>xins. Microbiol Rev 51:206–220, 1987.O’Brien, J.D. Swollen head syndrome in broiler breeders. Vet Rec 117:619–620, 1985.Pages Mante, A., L. Costa Quintana. Síndrome de cabeza hinchada (SH). Etiología <strong>and</strong> profilaxis.Med Vet 4:53–57, 1987.Riley, L.W., R.S. Remis, S.D. Helgerson, et al. Hemorrhagic colitis associated with a rareEscherichia coli serotype. N Engl J Med 308:681–685, 1983.


CORYNEBACTERIOSIS 99Robins-Browne, R.M., M.M. Levine, B. Rowe, E.M. Gabriel. Failure <strong>to</strong> detect conventionalentero<strong>to</strong>xins in classical enteropathogenic (serotyped) Escherichia coli strains of proven pathogenicity.Infect Immun 38:798–801, 1982.Rowe, B., J. Taylor, K.A. Bettelheim. An investigation of traveller’s diarrhoea. Lancet1:1–5, 1970.Rutter, J.M., G.W. Jones, G.T. Brown, M.R. Burrows, P.D. Luther. Antibacterial activity incolostrum <strong>and</strong> milk associated with protection of piglets against enteric disease caused byK88-positive Escherichia coli. Infect Immun 13:667–676, 1976.Ryder, R.W., R.A. Kaslow, J.G. Wells. Evidence for entero<strong>to</strong>xin production by a classicenteropathogenic serotype of Escherichia coli. J Infect Dis 140:626–628, 1979.Saltys, M.A. Bacteria <strong>and</strong> Fungi Pathogenic <strong>to</strong> Man <strong>and</strong> Animals. London: Baillière,Tindall <strong>and</strong> Cox; 1963.Spencer, L. Escherichia coli O157:H7 infection forces awareness of food production <strong>and</strong>h<strong>and</strong>ling. J Am Vet Med Assoc 202:1043–1047, 1993.Timoney, J.F., J.H. Gillespie, F.W. Scott, J.E. Barlough. Hagan <strong>and</strong> Bruner’s Microbiology<strong>and</strong> Infectious Diseases of Domestic Animals. 8th ed. Ithaca: Coms<strong>to</strong>ck; 1988.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Preliminary report: Foodborne outbreak of Escherichia coliO157:H7 infections from hamburgers—western United States, 1993. MMWR Morb MortWkly Rep 42:85–86, 1993.Vial, P.A., R. Robins-Browne, H. Lior, et al. Characterization of enteroadherent-aggregativeEscherichia coli, a putative agent of diarrheal disease. J Infect Dis 158:70–79, 1988.White, D.G., R.A. Wilson, A. San Gabriel, et al. Genetic relationship among strainsof avian Escherichia coli associated with swollen-head syndrome. Infect Immun 58:3613–3620, 1990.Williams, L.P., B.C. Hobbs. Enterobacteriaceae infections. In: Hubbert, W.T., W.F.McCulloch, P.R. Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed.Springfield: Thomas; 1975.World Health Organization (<strong>WHO</strong>). Research priorities for diarrhoeal disease vaccines:Memor<strong>and</strong>um from a <strong>WHO</strong> meeting. Bull <strong>WHO</strong> 69:667–676, 1991.World Health Organization Scientific Working Group. Escherichia coli diarrhoea. Bull<strong>WHO</strong> 58:23–36, 1980.CORYNEBACTERIOSISICD-10 A48 other bacterial <strong>diseases</strong>, not elsewhere classifiedEtiology: The genus Corynebacterium consists of slightly inflexed, gram-positive,non–acid-fast, nonmotile, nonsporogenic, nonencapsulated, facultatively aerobicor anaerobic, catalase-positive bacilli. The genus is related <strong>to</strong> Nocardia,Rhodococcus, <strong>and</strong> Mycobacterium.The genus Corynebacterium includes species such as C. diphtheriae (typespecies), the agent of human diphtheria, <strong>and</strong> such animal pathogens as C. pseudotuberculosis(C. bovis) <strong>and</strong> C. renale. There are also species that are pathogenic for


100 BACTERIOSESplants <strong>and</strong> others that are saprophytes. Corynebacteria, with the exception of thespecies C. diphtheriae, are often called diphtheroids.The species that are animal commensals or pathogens <strong>and</strong> are transmitted <strong>to</strong> manare C. pseudotuberculosis, C. ulcerans, C. bovis (the latter two are still not recognizedas species), C. kutscheri, <strong>and</strong> a group of three species: C. renale, C. pilosum,<strong>and</strong> C. cystitidis.Geographic Distribution: Worldwide.Occurrence in Man: Few cases have been recognized.Occurrence in Animals: C. pseudotuberculosis (C. ovis) occurs in many parts ofthe world among sheep <strong>and</strong> goats. It is less frequent in horses <strong>and</strong> camels. C. bovisis a commensal bacteria in the udder <strong>and</strong> genital tract of bovines. It may occasionallycause mastitis (Gillespie <strong>and</strong> Timoney, 1981). C. ulcerans is found in the nose<strong>and</strong> throat of man <strong>and</strong> horses (Wiggins et al., 1981). The species of the group C.renale are frequent etiological agents of cystitis, ureteritis, <strong>and</strong> pyelonephritis inbovines. C. kutscheri is a commensal <strong>and</strong> pathogen in rodents.The Disease in Man: Twelve human cases caused by C. pseudotuberculosis (C.ovis) have been described. The <strong>common</strong> lesion in these patients was a suppurativegranuloma<strong>to</strong>us lymphadenitis. There was only one different clinical picture: a veterinarystudent who contracted eosinophilic pneumonia after exposure in a microbiologylabora<strong>to</strong>ry. The victims were treated with erythromycin or tetracycline forseveral weeks (Brown, 1990). Almost all strains of C. pseudotuberculosis produce adermonecrotic <strong>to</strong>xin.C. ulcerans has caused a variety of pathological symp<strong>to</strong>ms in man, particularlypharyngitis, but also ulcers in the limbs, presumed cases of pneumonia, <strong>and</strong> a <strong>diseases</strong>imilar <strong>to</strong> diphtheria, with pseudomembranes <strong>and</strong> cardiac <strong>and</strong> neurologicalmanifestations (Brown, 1990; Krech <strong>and</strong> Hollis, 1991).C. bovis is a <strong>common</strong> commensal in cow’s milk, whose fat it hydrolyzes. The literaturedescribes seven human cases of disease caused by this agent. Three of thesehad CNS impairment <strong>and</strong> the others had prosthetic valve endocarditis, chronic otitis,<strong>and</strong> a persistent ulcer on one leg (Vale <strong>and</strong> Scott, 1977; Brown, 1990).C. renale has caused rectal <strong>and</strong> chest abscesses.C. kutscheri is an opportunistic pathogen in wild <strong>and</strong> labora<strong>to</strong>ry rodents (rats <strong>and</strong>mice). There are only two known human cases of disease caused by this agent: onewith septic arthritis <strong>and</strong> the other a premature infant with chorioamnionitis (Krech<strong>and</strong> Hollis, 1991). The species is not clearly defined in the human cases describedin the literature.The recommended treatment is simultaneous administration of rifampicin <strong>and</strong>erythromycin (Brown, 1990).The Disease in Animals: The corynebacterioses are much more important in veterinarymedicine. Some of the <strong>diseases</strong> are described briefly below (Timoney et al., 1988).C. pseudotuberculosis is the usual etiologic agent of caseous lymphadenitis insheep <strong>and</strong> goats, which occurs in many parts of the world where these animalspecies are raised. The agent gains entry through wounds <strong>and</strong> localizes in theregional lymph nodes, where it forms a caseous greenish pus. Abscesses may alsobe found in the lungs, as well as in the mediastinal <strong>and</strong> mesenteric lymph nodes.


CORYNEBACTERIOSIS 101Two different pathological conditions have been found in horses. One is ulcerativelymphangitis, with metacarpal <strong>and</strong> metatarsophalangeal abscesses that containa thick, greenish pus <strong>and</strong> at times leave an ulceration that is slow <strong>to</strong> heal. The otherconsists of large <strong>and</strong> painful abscesses on the chest <strong>and</strong> in the inguinal <strong>and</strong> abdominalregions. It may also affect camels, deer, mules, <strong>and</strong> bovines.C. pseudotuberculosis has two serotypes. Serotype 1 predominates in sheep <strong>and</strong>goats, <strong>and</strong> serotype 2 in buffalo <strong>and</strong> cows. It produces an exo<strong>to</strong>xin, phospholipaseD, which gives the bacteria much of its virulence by increasing vascular permeability.The other virulence fac<strong>to</strong>rs are a thermostable pyogenous fac<strong>to</strong>r that attractsleukocytes <strong>and</strong> a surface lipid that is <strong>to</strong>xic <strong>to</strong> leukocytes.C. renale is the most frequent agent in the group that causes pyelonephritis. It isalso responsible for many cases of cystitis <strong>and</strong> ureteritis, particularly in cows. Thisbacteria produces diphtherial inflammation of the bladder, ureters, kidneys, <strong>and</strong>pelvis. It can be found in healthy cows in herds with sick animals. C. renale alsoaffects horses <strong>and</strong> sheep sporadically. C. pilosum is not very virulent <strong>and</strong> is onlyoccasionally the agent of pyelonephritis. C. cystitidis causes severe hemorrhagiccystitis, followed by pyelonephritis. C. bovis is usually a commensal in the udder<strong>and</strong> is only sometimes the primary agent of mastitis.C. ulcerans is a commensal in bovines <strong>and</strong> horses. It has been isolated frommilk <strong>and</strong> is presumed <strong>to</strong> occasionally cause mastitis in cows (Lipsky et al., 1982).An outbreak of gangrenous dermatitis caused by C. ulcerans occurred inRichardson ground squirrels (Spermophilus richardsonii) captured within the citylimits of Calgary (Canada). Between two <strong>and</strong> five months after capture, 63 (18%)of the animals fell ill with symp<strong>to</strong>ms of dermatitis <strong>and</strong> cellulitis. Some of the 350squirrels captured died, probably due <strong>to</strong> <strong>to</strong>xemia <strong>and</strong>/or septicemia, <strong>and</strong> had lesionsfrom acute necrotic dermatitis over a large part of their bodies. Pharyngitis wasfound in 4 of the 10 that were examined (Olson et al., 1988). The infection isassumed <strong>to</strong> have spread through bites, in a manner similar <strong>to</strong> that described inmonkeys (May, 1972).Most infections due <strong>to</strong> C. kutscheri in rodents are subclinical. Clinical cases shownasal <strong>and</strong> ocular secretion, as well as dyspnea, arthritis, <strong>and</strong> cutaneous abscesses thatform gray nodules some 15 mm in diameter. Upon au<strong>to</strong>psy, abscesses are found inthe liver, kidneys, lungs, <strong>and</strong> lymph nodes. Diagnosis can be performed through culture<strong>and</strong> isolation of the etiologic agent or serology (ELISA, complement fixation,agglutination). Treatment with penicillin can prevent the appearance of clinicalsymp<strong>to</strong>ms in animals in an affected colony, but does not eliminate carrier status(Fraser et al., 1991).C. diphtheriae is an exclusively human pathogen. However, in an outbreak tha<strong>to</strong>ccurred in a colony of 300 guinea pigs in Nigeria, 60 died with pneumonia lesions,endometritis, <strong>and</strong> slight intestinal congestion. C. diphtheriae was considered thecause of death, since it was isolated from the lungs <strong>and</strong> heart blood. The source ofinfection could not be determined (Okewole et al., 1990).Treatment with high doses of penicillin is effective if begun early in the course ofthe disease.Source of Infection <strong>and</strong> Mode of Transmission: The corynebacteria describedhere are considered zoonotic, with the exception of C. diphtheriae, for which thereservoir is man <strong>and</strong> transmission is from human <strong>to</strong> human.


102 BACTERIOSESThe reservoir of C. pseudotuberculosis is sheep <strong>and</strong> goats. Man acquires theinfection through contact with sick animals, their organs, or products (skins, milk).Among sheep <strong>and</strong> goats, the infection is transmitted from an animal with an openabscess <strong>to</strong> another animal with abrasions, such as those produced during shearing.Sometimes C. pseudotuberculosis can penetrate through abrasions in the oralmucosa, or it can be inhaled <strong>and</strong> cause abscesses in the lungs (Timoney et al., 1988).C. ulcerans is a <strong>common</strong> commensal in bovines <strong>and</strong> horses. The bacteria is probablytransmitted <strong>to</strong> man through raw milk. The infection may also be transmitted viathe airborne route (Brown, 1990).C. bovis is a commensal in the reproductive system of bovines <strong>and</strong> can frequentlybe found in milk; only occasionally does it cause mastitis. In a survey conducted in74 dairy farms in Ontario (Canada), C. bovis was found in the milk of 36% of thecows (Brooks et al., 1983).The reservoir of C. renale, C. pilosum, <strong>and</strong> C. cystitidis is bovines. The mode oftransmission from bovines <strong>to</strong> man is unclear. C. renale <strong>and</strong> C. pilosum are transmittedamong bovines when the urine from a sick cow reaches the vulva of a healthy cow. C.cystitidis is a commensal of the prepuce of bulls <strong>and</strong> can be transmitted sexually. It canalso be transmitted when drops of urine are sprinkled from one cow <strong>to</strong> another.Diagnosis: A diagnosis of human corynebacteriosis can only be confirmedthrough isolation <strong>and</strong> identification of the species.The same applies <strong>to</strong> animal corynebacteriosis, although in the case of caseouslymphadenitis in the surface lymph nodes of sheep <strong>and</strong> goats, the lesions, along witha gram-stained smear, are sufficiently characteristic for diagnosis. Several serologicaltests have been used <strong>to</strong> detect healthy carriers of C. pseudotuberculosis.Control: The few human cases identified <strong>to</strong> date do not justify the establishmen<strong>to</strong>f special preventive measures. However, correct diagnosis is important for effectivetreatment.To prevent caseous lymphadenitis due <strong>to</strong> C. pseudotuberculosis, it is essential <strong>to</strong>avoid lesions during shearing. When they do occur, they should be treated promptly<strong>and</strong> correctly.BibliographyBrooks, B.W., D.A. Barnum, A.H. Meek. An observational study of Corynebacterium bovisin selected Ontario dairy herds. Can J Comp Med 47:73–78, 1983.Brown, A.E. Other corynebacteria. In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E. Bennett, eds.Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: Churchill Livings<strong>to</strong>ne, Inc.;1990.Fraser, C.M., J.A. Bergeron, A. Mays, S.E. Aiello. The Merck Veterinary Manual. 7th ed.Rahway: Merck; 1991.Gillespie, J.H., J.F. Timoney. Hagan <strong>and</strong> Bruner’s Infectious Diseases of DomesticAnimals. 7th ed. Ithaca: Coms<strong>to</strong>ck; 1981.Krech, T., D.G. Hollis. Corynebacterium <strong>and</strong> related organisms. In: Balows, A., W.J.Hausler, K.L. Hermann, H.D. Isenberg, H.J. Shadomy, eds. Manual of Clinical Microbiology.5th ed. Washing<strong>to</strong>n, D.C.: American Society for Microbiology; 1991.Lipsky, B.A., A.C. Goldberger, L.S. Tompkins, J.J. Plorde. Infections caused by nondiphtheriacorynebacteria. Rev Infect Dis 4:1220–1235, 1982. Cited in: Krech, T., D.G. Hollis.


DERMATOPHILOSIS 103Corynebacterium <strong>and</strong> related organisms. In: Balows, A., W.J. Hausler, K.L. Hermann, H.D.Isenberg, H.J. Shadomy, eds. Manual of Clinical Microbiology. 5th ed. Washing<strong>to</strong>n, D.C.:American Society for Microbiology; 1991.May, B.D. Corynebacterium ulcerans infections in monkeys. Lab Anim Sci 22:509–513, 1972.Meers, P.D. A case of classical diphtheria <strong>and</strong> other infections due <strong>to</strong> Corynebacteriumulcerans. J Infect 1:139–142, 1979.Okewole, P.A., O.S. Odeyemi, E.A. Irokanulo, et al. Corynebacterium diphtheriae isolatedfrom guinea pigs. Indian Vet J 67:579–580, 1990.Olson, M.E., I. Goemans, D. Bolingbroke, S. Lundberg. Gangrenous dermatitis caused byCorynebacterium ulcerans in Richardson ground squirrels. J Am Vet Med Assoc 193:367–368,1988.Rountree, P.M., H.R. Carne. Human infection with an unusual corynebacterium. J PatholBacteriol 94:19–27, 1967.Timoney, J.F., J.H. Gillespie, F.W. Scott, J.E. Barbough. Hagan <strong>and</strong> Bruner’s Microbiology<strong>and</strong> Infectious Diseases of Domestic Animals. 8th ed. Ithaca: Coms<strong>to</strong>ck; 1988.Vale, J.A., G.W. Scott. Corynebacterium bovis as a cause of human disease. Lancet2:682–684, 1977.Van Etta, L.L., G.A. Filice, R.M. Ferguson, D.N. Gerding. Corynebacterium equi:A reviewof 12 cases of human infection. Rev Infect Dis 5:1012–1018, 1983.Wiggins, G.L., F.O. Sottnek, G.Y. Hermann. Diphtheria <strong>and</strong> other corynebacterial infections.In: Balows, A., W.J. Hausler, Jr., eds. Diagnostic Procedures for Bacterial, Mycotic <strong>and</strong>Parasitic Infections. 6th ed. Washing<strong>to</strong>n, D.C.: American Public Health Association; 1981.Willet, H.P. Corynebacterium. In: Joklik, W.K., H.P. Willet, D.B. Amos, eds. ZinsserMicrobiology. 17th ed. New York: Apple<strong>to</strong>n-Century-Crofts; 1980.


DERMATOPHILOSISICD-10 A48.8 other specified bacterial <strong>diseases</strong>Synonyms: Strep<strong>to</strong>thrichosis, mycotic dermatitis (in sheep).Etiology: Derma<strong>to</strong>philus congolensis (D. derma<strong>to</strong>nomus, D. pedis) is a bacteriumbelonging <strong>to</strong> the order Actinomycetales. It is facultatively anaerobic, grampositive,<strong>and</strong> non–acid-fast. D. congolensis is characterized by branched filamentswith transverse <strong>and</strong> longitudinal septation. When the filaments mature, they fragment<strong>and</strong> release motile, flagellate spores, called zoospores, which constitute theinfective agent. In turn, the zoospores germinate <strong>and</strong> form filaments that producenew zoospores, thus repeating the cycle.Geographic Distribution: Worldwide. Derma<strong>to</strong>philosis has been confirmed inmany areas of Africa, Australia, Europe, <strong>and</strong> New Zeal<strong>and</strong>, as well as in North <strong>and</strong>South America.Occurrence in Man: The first known cases were identified in 1961 in New York(USA), where four people became ill after h<strong>and</strong>ling a deer with derma<strong>to</strong>philosis


104 BACTERIOSESlesions. Subsequently, several other cases were described: one in a student at theUniversity of Kansas (USA), three cases in Australia, <strong>and</strong> two in Brazil (Kaplan,1980; Portugal <strong>and</strong> Baldassi, 1980). A case was recorded in Costa Rica of a veterinarianwho came in<strong>to</strong> contact with infected cattle.Occurrence in Animals: The disease has been observed in several species ofdomestic <strong>and</strong> wild animals. Those most frequently affected are cattle, sheep, goats,<strong>and</strong> horses. The disease is most prevalent in tropical <strong>and</strong> subtropical climates. Theimportance of derma<strong>to</strong>philosis lies in the economic losses it causes, due <strong>to</strong> the damage<strong>to</strong> leather, wool, <strong>and</strong> pelts. In some African countries, from 16% (Kenya) <strong>to</strong> 90%(Tanzania) of cow hides have been damaged. In Great Britain, it has been estimatedthat affected fine wool loses 20% of its commercial value. Moreover, shearing is difficultin chronically sick woolbearing animals.The Disease in Man: In the few known cases, the disease has been characterizedby pimples <strong>and</strong> multiple pustules (2–25) on the h<strong>and</strong>s <strong>and</strong> forearms, containing aserous or yellowish white exudate. Upon rupturing, they left a reddish crateriformcavity. The lesions healed in 3 <strong>to</strong> 14 days, leaving a purplish red scar.The Disease in Animals: In derma<strong>to</strong>philosis or strep<strong>to</strong>trichosis in bovines, sheep,horses, or goats, a serous exudate at the base of hair tufts dries <strong>and</strong> forms a scab.When the scab comes off, it leaves a moist alopecic area. The lesions vary in size;some may be very small <strong>and</strong> go unnoticed, but at times they are confluent <strong>and</strong> covera large area. In general, they are found on the back, head, neck, <strong>and</strong> places whereticks attach. In sheep, the disease known as mycotic dermatitis (lumpy wool) beginswith hyperemia <strong>and</strong> swelling of the affected area of skin, <strong>and</strong> an exudation thatbecomes hard <strong>and</strong> scablike. In chronic cases, conical hard crusts with a horny consistencyform around tufts of wool. In mild cases, the disease is seen only duringshearing, since it makes the operation difficult. Animals do not experience a burningsensation <strong>and</strong> are not seen <strong>to</strong> scratch themselves against posts or other objects.Secondary infections may cause death in lambs. Derma<strong>to</strong>philosis is also a fac<strong>to</strong>rfavoring semispecific myiases (see section on myiases in Volume III: ParasiticDiseases), caused in Australia by Lucillia cuprina (the principal agent of “bodystrike”). The fly not only prefers the moist areas affected by derma<strong>to</strong>philosis aboveother moist areas in the fur for egg laying, but larval development is aided by theskin lesion caused by D. congolensis (Gherardi et al., 1981).In Great Britain, a localized form of the disease in the distal regions of the extremitiesof sheep has been confirmed <strong>and</strong> named proliferative hoof dermatitis. This formis characterized by extensive inflammation of the skin <strong>and</strong> formation of thick scabs.The scabs come loose, revealing small hemorrhagic dots that cause the lesion <strong>to</strong>resemble a strawberry, from which the disease’s <strong>common</strong> name, “strawberry footrot,” is derived. In cases without complications <strong>and</strong> in the dry season, the lesionsheal spontaneously in about three weeks.In derma<strong>to</strong>philosis cases described in domestic cats, the lesions differ from thoseof other domestic species in that they affect deeper tissues. In cats, granuloma<strong>to</strong>uslesions due <strong>to</strong> D. congolensis have been found on the <strong>to</strong>ngue, bladder, <strong>and</strong> popliteallymph nodes (Kaplan, 1980).Source of Infection <strong>and</strong> Mode of Transmission: The etiologic agent, D. congolensis,is an obligate parasite that has been isolated only from lesions in animals.


DERMATOPHILOSIS 105However, according <strong>to</strong> Bida <strong>and</strong> Dennis (1977), the agent can be found in the soilduring the dry season.Environmental humidity <strong>and</strong> moist skin are predisposing fac<strong>to</strong>rs in the disease. Thezoospore needs moisture <strong>to</strong> mobilize <strong>and</strong> be released. The rainy seasons in tropical climatesare the most favorable <strong>to</strong> spread of the infection. Another important fac<strong>to</strong>r insheep is malnutrition, which usually occurs during the dry season due <strong>to</strong> the lack ofpasture. Malnourished animals have more persistent <strong>and</strong> chronic lesions than wellnourishedanimals. The difference is probably due <strong>to</strong> the reduced growth of wool <strong>and</strong>reduced production of lanoline in malnourished animals (S<strong>and</strong>ers et al., 1990). Mostresearchers assign great importance <strong>to</strong> the level of tick infestation in cattle (Koney <strong>and</strong>Morrow, 1990) <strong>and</strong> other animal species, as well as <strong>to</strong> infestation by other insects.Human cases have arisen from direct contact with animal lesions. Man is probablyquite resistant <strong>to</strong> the infection, as the number of human cases is small despite thefrequency of the disease in animals.The most <strong>common</strong> means of transmission between animals seems <strong>to</strong> be mechanicaltransport by arthropod vec<strong>to</strong>rs, including ticks, flies, <strong>and</strong> mosqui<strong>to</strong>es. The infectiveelement is the zoospore. Most infections occur at the end of spring <strong>and</strong> in summer,when insects are most abundant. An important fac<strong>to</strong>r in transmission ismoisture, which allows the zoospore <strong>to</strong> detach from the mycelium.The most serious outbreaks occur during prolonged humid seasons <strong>and</strong> during therainy season in tropical areas. Sheep with long wool that retains moisture are mostsusceptible <strong>to</strong> the infection. During dry seasons, the agent can survive in moist spotson the body, such as the axilla or in skinfolds.The infection may also be transmitted by means of objects, such as plant thornsor shears that cause lesions on the extremities or on the lips.Role of Animals in the Epidemiology of the Disease: The infection is transmittedfrom one animal <strong>to</strong> another <strong>and</strong> only occasionally from animal <strong>to</strong> man. The onlyknown reservoirs of the agent are domestic <strong>and</strong> wild animals.Diagnosis: Clinical diagnosis is confirmed by microscopic examination ofstained smears (Giemsa, methylene blue, or Wright’s stain) made from exudates orscabs. This is the simplest <strong>and</strong> most practical method. Immunofluorescence mayalso be used on smears or tissue samples.The isolation of the agent should be done in rich media such as blood agar. Thisculture method is often difficult due <strong>to</strong> contamination. To overcome this difficulty,passage through rabbits has been used.Several serological methods have been used <strong>to</strong> detect antibodies <strong>to</strong> D. congolensis.In a study comparing passive hemagglutination, immunodiffusion in agar gel,<strong>and</strong> counterimmunoelectrophoresis, the last test gave the best results in terms ofboth sensitivity <strong>and</strong> specificity (Makinde <strong>and</strong> Majiyagbe, 1982).Control: Given the few cases of derma<strong>to</strong>philosis in man, special control measures<strong>to</strong> protect against infection are not justified. Nevertheless, it would be prudent not <strong>to</strong>h<strong>and</strong>le animals with lesions with bare h<strong>and</strong>s (especially if one has abrasions or skinwounds).In Africa, tick control has been shown <strong>to</strong> be effective in preventing bovine derma<strong>to</strong>philosis.Sheep with mycotic dermatitis should be shorn last or, preferably, in a separateplace. Affected wool should be burned. Satisfac<strong>to</strong>ry results have been obtained using


106 BACTERIOSES1% alum dips. In chronic cases, an intramuscular injection of 70 mg of strep<strong>to</strong>mycin<strong>and</strong> 70,000 units of penicillin may be administered two months before shearing.This drug therapy seems <strong>to</strong> be very effective <strong>and</strong> prevents difficulties in shearing.The use of antibiotics (strep<strong>to</strong>mycin, penicillin, <strong>and</strong> others) was effective in producingclinical cure or improvement in affected animals, but did not always eliminatethe causal agent.The infection is controlled by isolating or eliminating chronically sick animals<strong>and</strong> combating ec<strong>to</strong>parasites. Externally applied insecticides are used <strong>to</strong> combat bitinginsects.The study of a vaccine against animal derma<strong>to</strong>philosis is in an experimental stage(Sutherl<strong>and</strong> <strong>and</strong> Robertson, 1988; How et al., 1990).BibliographyAinsworth, G.C., P.K.C. Austwick. Fungal Diseases of Animals. 2nd ed. Farnham Royal,Slough, United Kingdom: Commonwealth Agriculture Bureau; 1973.Bida, S.A., S.M. Dennis. Sequential pathological changes in natural <strong>and</strong> experimental derma<strong>to</strong>philosisin Bunaji cattle. Res Vet Sci 22:18–22, 1977.Carter, G.R. Diagnostic Procedures in Veterinary Microbiology. 2nd ed. Springfield:Thomas; 1973.Dean, D.J., M.A. Gordon, C.W. Sveringhaus, E.T. Kroll, J.R. Reilly. Strep<strong>to</strong>thricosis: Anew zoonotic disease. N Y State J Med 61:1283–1287, 1961.Gherardi, S.G., N. Monzu, S.S. Sutherl<strong>and</strong>, K.G. Johnson, G.M. Robertson. The associationbetween body strike <strong>and</strong> derma<strong>to</strong>philosis of sheep under controlled conditions. Aust Vet J57:268–271, 1981.Gordon, M.A. The genus Derma<strong>to</strong>philus. J Bacteriol 88:508–522, 1964.How, S.J., D.H. Lloyd, A.B. S<strong>and</strong>ers. Vaccination against Derma<strong>to</strong>philus congolensis infectionin ruminants: Prospects for control. In: Tcharnev C., R. Halliwell, eds. Advances inVeterinary Derma<strong>to</strong>logy. London: Baillière Tindall; 1990.Kaplan, W. Derma<strong>to</strong>philosis in man <strong>and</strong> lower animals: A review. In: Proceedings of theFifth International Conference on the Mycoses. Superficial, cutaneous, <strong>and</strong> subcutaneousinfections. Caracas, Venezuela, 27–30 April 1980. Washing<strong>to</strong>n, D.C.: Pan American HealthOrganization; 1980. (Scientific Publication 396).Koney, E.B.M., A.N. Morrow. Strep<strong>to</strong>thricosis in cattle on the coastal plains of Ghana: Acomparison of the disease in animals reared under two different management systems. TropAnim Health Prod 22:89–94, 1990.Makinde, A.A., K.A. Majiyagbe. Serodiagnosis of Derma<strong>to</strong>philus congolensis infection bycounterimmunoelectrophoresis. Res Vet Sci 33:265–269, 1982.Pier, A.C. Géneros Actinomyces, Nocardia y Derma<strong>to</strong>philus. In: Merchant, I.A., R.A.Packer. Bacteriología veterinaria. 3.ª ed. Zaragoza, España: Acribia; 1970.Portugal, M.A.C.S., L. Baldassi. A derma<strong>to</strong>filose no Brasil. Revisão bibliográfica. Arq InstBiol 47:53–58, 1980.Roberts, D.S. Derma<strong>to</strong>philus infection. Vet Bull 37:513–521, 1967.S<strong>and</strong>ers, A.B., S.J. How, D.H. Lloyd, R. Hill. The effect of energy malnutrition in ruminantson experimental infection with Derma<strong>to</strong>philus congolensis. J Comp Pathol103:361–368, 1990.Sutherl<strong>and</strong>, S.S., G.M. Robertson. Vaccination against ovine derma<strong>to</strong>philosis. Vet Microbiol18:285–295, 1988.


DISEASES CAUSED BY NONTUBERCULOUS MYCOBACTERIA 107DISEASES CAUSED BY NONTUBERCULOUSMYCOBACTERIAICD-10 A31.0 pulmonary mycobacterial infection;A31.1 cutaneous mycobacterial infection;A31.8 other mycobacterial infectionsSynonyms: Mycobacteriosis, nontuberculous mycobacteriosis, nontuberculousmycobacterial infection.Etiology: The etiologic agents of nontuberculous mycobacteriosis (NTM) form agroup separate from those that cause tuberculosis in mammals, Mycobacteriumtuberculosis, M. bovis, M. africanum, <strong>and</strong> M. microti (the agent of tuberculosis inrodents). Previously called anonymous, atypical, or unclassified mycobacteria, theyhave since been characterized <strong>and</strong> given specific names.Mycobacteria potentially pathogenic for man <strong>and</strong> animals currently include some15 species. The most important group among these species is Mycobacterium aviumcomplex (MAC), replacing what was formerly called MAI (M. avium-intracellulare)or MAIS (M. avium-intracellulare-scrofulaceum). These mycobacteriaare important pathogens for birds (avian tuberculosis) <strong>and</strong> some mammals (swinetuberculosis). MAC has become important as a human pathogen due <strong>to</strong> theAIDS epidemic.There are both genetic <strong>and</strong> antigenic indications that M. paratuberculosis, theagent of chronic hypertrophic enteritis in cattle <strong>and</strong> sheep, should be included in thesame complex as M. avium (Grange et al., 1990). There are also data suggesting thatthe mycobacterial strains isolated from patients with Crohn’s disease are geneticallyrelated <strong>to</strong> M. paratuberculosis (S<strong>and</strong>erson et al., 1992).M. paratuberculosis is characterized by its requirement of mycobactin (a lipid thatbinds iron) for growth in culture media. There are also strains similar <strong>to</strong> MAC thatare mycobactin-dependent <strong>to</strong> a greater or lesser degree, among them the strains isolatedfrom the wild pigeon (Palumba palumbus), which through experimental inoculationin cattle produces a disease similar <strong>to</strong> paratuberculosis.DNA:DNA hybridization studies demonstrated that M. avium, M. paratuberculosis,<strong>and</strong> the mycobacteria of the European wild pigeon (Palumba palumbus) belong<strong>to</strong> a single genomic species. Based on numerical taxonomy studies of mycobactindependentmycobacteria, DNA sequences, <strong>and</strong> genotype <strong>and</strong> other studies, Thorel etal. (1990) suggest dividing the species in<strong>to</strong> M. avium subsp. avium, M. avium subsp.paratuberculosis, <strong>and</strong> M. avium subsp. silvaticum. The latter would correspond <strong>to</strong>the mycobacteria isolated from the wild pigeon.MAC is composed of 28 serotypes (1–28); the first three belong <strong>to</strong> M. avium, <strong>and</strong>the rest <strong>to</strong> M. intracellulare. Serotyping has been valuable in research but is notapplicable in routine labora<strong>to</strong>ries <strong>and</strong> has been discontinued. Runyon’s classification,developed in 1959, is still in use. It subdivides the mycobacteria in<strong>to</strong> four largegroups: pho<strong>to</strong>chromogens (Group 1), sco<strong>to</strong>chromogens (Group 2), nonchromogens(Group 3), <strong>and</strong> rapid growers (Group 4). The different species of mycobacteria aredistinguished by their phenotypic characteristics, such as optimum growth temperature,rapid or slow growth, utilization of niacin, nitrate reduction, <strong>and</strong> other biochemicalproperties (Wayne <strong>and</strong> Kubica, 1986).


108 BACTERIOSESThe mycobacteria that are potentially pathogenic for man <strong>and</strong> animals include theslow-growing MAC, M. kansasii, M. marinum, M. xenopi, M. szulgai, <strong>and</strong> M.simiae; <strong>and</strong> the fast-growing M. fortuitum <strong>and</strong> M. chelonae (or M. fortuitumcomplex).Geographic Distribution: Their presence, distribution, <strong>and</strong> relative importanceas a cause of disease have been studied primarily in the more developed countries,where the prevalence of tuberculosis is also lower. Some species are distributedworldwide, while others predominate in certain areas. For example, the pulmonarydisease in man caused by M. kansasii is prevalent in Engl<strong>and</strong> <strong>and</strong> Wales (UnitedKingdom), <strong>and</strong> in Kansas City, Chicago, <strong>and</strong> the state of Texas (USA). On the otherh<strong>and</strong>, the disease caused by MAC is more frequent in the southeastern United States,western Australia, <strong>and</strong> Japan (Wolinsky, 1979). The situation has changed radicallywith the advance of the AIDS epidemic.Distribution is similar in animals, since the infection comes from an environmentalsource. These agents are believed <strong>to</strong> be more important in hot <strong>and</strong> humid areasthan in temperate <strong>and</strong> cold climates.Occurrence in Man: A distinction must be made between colonization <strong>and</strong> temporarysensitivity, infection, <strong>and</strong> cases of disease. Since diagnosis depends on theisolation <strong>and</strong> typing of the etiologic agent, most confirmations come from countrieswith a good system of labora<strong>to</strong>ries. In Australia, the annual rate of pulmonary infectionhas been estimated at 1.7 <strong>to</strong> 4 cases per 100,000 inhabitants in Queensl<strong>and</strong> <strong>and</strong>from 0.5 <strong>to</strong> 1.2 per 100,000 in the entire country. In the Canadian province of BritishColumbia, the annual rate for all nontuberculous mycobacterial <strong>diseases</strong> increasedfrom 0.17 <strong>to</strong> 0.53 per 100,000 inhabitants between 1960 <strong>and</strong> 1972 (Wolinsky, 1979).The incidence of MAC in AIDS patients continues <strong>to</strong> increase. In the US, it was5.7% in the period 1985–1988, while it reached 23.3% in 1989–1990 (Havlik et al.,1992). Isolates of nontuberculous mycobacteria from 727 AIDS patients in the US(sample from the entire country) were sent <strong>to</strong> the Centers for Disease Control <strong>and</strong>Prevention for serotyping. It was possible <strong>to</strong> type 87% <strong>and</strong> almost all the isolatesbelonged <strong>to</strong> MAC serotypes 1 <strong>to</strong> 6 <strong>and</strong> 8 <strong>to</strong> 11. Most M. avium isolates <strong>and</strong> the isolatesthat could not be typed were taken from blood samples. M. intracellulare madeup of only 3% of the isolates. More than 50% of all the cultures came from NewYork <strong>and</strong> California (Yakrus <strong>and</strong> Good, 1990).A prospective study of AIDS patients was able <strong>to</strong> diagnose MAC only in thosewho had a CD4+ count of less than 100 cells/mm 3 . These patients had fever, diarrhea,<strong>and</strong> weight loss (Havlik et al., 1992).In Zurich (Switzerl<strong>and</strong>), a retrospective study covering the period 1983–1988examined patients negative for human immunodeficiency virus (HIV).Nontuberculous mycobacteria were isolated from 513 cases, 34 of whom had anobvious disease. In 23 of the 34 cases, the disease was pulmonary; the soft tissueswere affected in 10 cases <strong>and</strong> there was 1 case of disseminated infection (Debrunneret al., 1992).In Argentina, 8,006 cultures from 4,894 patients were studied. Of these cultures,113 (1.4%) were identified as nontuberculous mycobacteria, belonging <strong>to</strong> 18 cases(0.37% of the <strong>to</strong>tal number of patients). The agents isolated were M. kansasii ineight cases, MAIS in another eight cases, M. marinum in one case, <strong>and</strong> an infectioncaused by both M. tuberculosis <strong>and</strong> M. kansasii in another case. Localization was


DISEASES CAUSED BY NONTUBERCULOUS MYCOBACTERIA 109pulmonary in 16 cases <strong>and</strong> cutaneous in 2 (Di Lonardo et al., 1983). A study conductedby 15 labora<strong>to</strong>ries in 6 regions of Argentina obtained 13,544 mycobacteriacultures from 7,662 patients. The etiologic agent was Mycobacterium tuberculosisin 99.17% of the patients, M. bovis in 0.47%, <strong>and</strong> MAIS in 0.35% (Barrera <strong>and</strong> DeKan<strong>to</strong>r, 1987). Between June 1985 <strong>and</strong> December 1991, at the Muñiz Hospital (forinfectious <strong>diseases</strong>) in Buenos Aires, the prevalence of nontuberculous mycobacterial<strong>diseases</strong> was 6.2% in HIV-positive or AIDS patients, <strong>and</strong> 0.5% in HIV-negativepatients (Di Lonardo et al., 1993).In Mexico, 547 cultures were made from samples taken from patients diagnosedwith tuberculosis using bacterioscopy. Of these cultures, 89.6% were identified asM. tuberculosis <strong>and</strong> 8.9% as potentially pathogenic nontuberculous mycobacteria,such as M. fortuitum, M. chelonae, M. scrofulaceum, <strong>and</strong> M. kansasii.Occurrence in Animals: The same considerations that apply <strong>to</strong> man arealso valid for animals. The disease has been confirmed in many mammalian <strong>and</strong>avian animal species, as well as poikilotherms. Among domestic animals, the diseaseis economically important in swine due <strong>to</strong> the losses it causes. MAC serotypes1 <strong>and</strong> 2 are the most <strong>common</strong>ly isolated from swine. These two serotypes are alsoresponsible for avian tuberculosis. Serotype 8 is an important pathogen for both man<strong>and</strong> animals (Thoen et al., 1981). Serotypes 4 <strong>and</strong> 5 are also isolated from swine inthe US.Surveillance <strong>and</strong> identification of mycobacteriosis in animals is mainly carriedout in countries where bovine tuberculosis has been controlled, as in the UnitedStates. Nontuberculous mycobacteria may interfere in the diagnosis of tuberculosis,causing unnecessary losses due <strong>to</strong> the slaughter of nontuberculous animals. There islittle information on animal mycobacteriosis in other areas.The Diseases in Man: The most <strong>common</strong> <strong>diseases</strong> in people with intact cellularimmunity are: (a) pulmonary disease, (b) lymphadenitis, <strong>and</strong> (c) soft tissue lesions.Other organs <strong>and</strong> tissues may be affected <strong>and</strong>, in some cases, hema<strong>to</strong>genous disseminationoccurs (Wolinsky, 1979).a) Chronic pulmonary disease resembling tuberculosis is the most important clinicalproblem caused by nontuberculous mycobacteria. The most <strong>common</strong> etiologicagents of this disease are MAC <strong>and</strong> M. kansasii. M. xenopi, M. scrofulaceum, M.szulgai, M. simiae, <strong>and</strong> M. fortuitum-chelonae are found less frequently. As withtuberculosis, there is great variation in the clinical presentation of the disease, fromminor lesions <strong>to</strong> an advanced disease with cavitation. Most cases occur in middleagedpersons who have preexisting pulmonary lesions (pneumoconiosis, chronicbronchitis, <strong>and</strong> others). Persons taking immunosuppressant drugs or with acquiredimmune deficiency are also susceptible. However, an appreciable percentage ofpatients have acquired the disease without having previous damage <strong>to</strong> the respira<strong>to</strong>ryor immune systems (Wolinsky, 1979).b) Mycobacterial lymphadenitis occurs in children from 18 months <strong>to</strong> 5 years ofage. The affected lymph nodes are primarily those of the neck close <strong>to</strong> the jaw bone,<strong>and</strong> generally on one side only. They soften rapidly <strong>and</strong> develop openings <strong>to</strong> the outside.The child’s general health is not affected. Calcification <strong>and</strong> fibrosis occur duringthe healing process.In countries at low risk for tuberculous infection, lymphadenitis due <strong>to</strong> MAC isprevalent, unlike countries with a medium <strong>to</strong> high prevalence of tuberculosis. In


110 BACTERIOSESBritish Columbia (Canada), the case rate was 0.37 per 100,000 inhabitants, while fortuberculous lymphadenitis due <strong>to</strong> M. tuberculosis, it was only 0.04 per 100,000inhabitants annually. In Great Britain, as in many other parts of the world, M. tuberculosisis prevalent in cases of lymphadenitis caused by Mycobacterium (Grange<strong>and</strong> Yates, 1990). The most <strong>common</strong> etiologic agents are different MAC serotypes,M. scrofulaceum, <strong>and</strong> M. kansasii. The proportion of each of these mycobacteriavaries by region. Other mycobacteria are isolated from lesions less frequently(Wolinsky, 1979).c) Diseases of the skin <strong>and</strong> subcutaneous tissue are caused by M. marinum, M.ulcerans, M. fortuitum, <strong>and</strong> M. chelonae.Localized abscesses ensue, particularly after injections, surgical interventions,war wounds, thorn penetration, <strong>and</strong> various traumas.Granulomas (swimming pool granuloma, fish tank granuloma) develop on theextremities as a group of papules that ulcerate <strong>and</strong> scab over. Lesions may persist formonths. Healing is usually spontaneous. The etiologic agent is M. marinum, whichinhabits <strong>and</strong> multiplies in fresh <strong>and</strong> salt water. M. marinum is a pho<strong>to</strong>chromogenalso found in marine animals; it grows well at 32°C <strong>and</strong> little or not at all at 37°C(S<strong>and</strong>ers <strong>and</strong> Horowitz, 1990). In Glenwood Spring, Colorado (USA), 290 cases ofgranuloma<strong>to</strong>us lesions were found among children who swam in a pool of tepidmineral water.Infections caused by M. ulcerans occur in many tropical areas of the world, particularlyin central Africa. They start as erythema<strong>to</strong>us nodules on the extremities <strong>and</strong>gradually become large, indolent ulcers with a necrotic base. This lesion is knownas “Buruli ulcer” in Africa <strong>and</strong> “Bairnsdale ulcer” in Australia.Infections caused by nontuberculous mycobacteria have also been described inthe joints, spinal column, <strong>and</strong> the urogenital tract, <strong>and</strong> as osteomyelitis of the sternumafter heart operations. A generalized, highly lethal infection may occur mainlyin leukemia patients or those undergoing treatment with immunosuppressants.Generalized infection with bacteremia detectable through hemoculture has beenconfirmed only in AIDS patients.Many other species of mycobacteria generally considered saprophytes can causepathological processes in man.There has been much interest <strong>and</strong> much controversy over the possibility that M.paratuberculosis, or a similar MAC mycobacteria, is the agent of Crohn’s disease.This chronic disease in man is of unknown etiology <strong>and</strong> causes a granuloma<strong>to</strong>usprocess in the terminal ileum, although lesions are also found in other parts of theintestine as well as the skin, the liver, <strong>and</strong> the joints. A mycobacterium that is theagent of chronic enteritis in cattle, sheep, <strong>and</strong> occasionally nonhuman primates (withcharacteristics very similar <strong>to</strong> M. paratuberculosis) was isolated from a few patients.It is a mycobactin-dependent mycobacterium that has biochemical, genomic, <strong>and</strong> culturingcharacteristics similar <strong>to</strong> M. paratuberculosis (except in the arylsulfatase <strong>and</strong>niacin reactions). It is experimentally pathogenic <strong>and</strong> capable of producing a granuloma<strong>to</strong>usdisease in the intestine of goats. Further research is needed <strong>to</strong> determinewhether this mycobacterium is actually the agent of Crohn’s disease (McFadden etal., 1987; Thorel, 1989; S<strong>and</strong>erson et al., 1992). A primary objective should be <strong>to</strong>improve culture media so that the mycobacterium can be isolated.Treatment of the pulmonary forms caused by MAC is difficult due <strong>to</strong> the resistanceof these mycobacteria <strong>to</strong> the antimicrobials <strong>common</strong>ly used in treating tuber-


DISEASES CAUSED BY NONTUBERCULOUS MYCOBACTERIA 111culosis. It is generally advisable <strong>to</strong> treat with various medications, selecting themafter conducting a sensitivity test on the isolated mycobacteria (Benenson, 1990).Such drugs would include isoniazid, rifampicin, <strong>and</strong> ethambu<strong>to</strong>l, adding strep<strong>to</strong>mycinat the start, <strong>and</strong> treatment must be sufficiently prolonged. Clarithromycin hasproven <strong>to</strong> be highly active in vitro <strong>and</strong> in vivo. The intracellular activity of clarithromycinincreases with ethambu<strong>to</strong>l <strong>and</strong> rifampicin. An evaluation of the variousdrugs can be found in the article by Inderlied et al. (1993). In cases of serious or disseminatedpulmonary disease, the patient may benefit from the addition of othermedications (S<strong>and</strong>ers <strong>and</strong> Horowitz, 1990). If the disease is limited—such as alocalized pneumopathy, a nodule, cervical lymphadenitis, or a subcutaneousabscess—surgical resection should be considered (Benenson, 1990).The Diseases in Animals: Many species of mammals <strong>and</strong> birds are susceptible <strong>to</strong>nontuberculous mycobacteria. The various MAC serotypes are the most importantetiological agents. The most frequent clinical form in mammals is lymphadenitis,but other tissues <strong>and</strong> organs may be affected (Thoen et al., 1981).CATTLE: In cattle, the most <strong>common</strong> nontuberculous mycobacterial infectionaffects the lymph gl<strong>and</strong>s. In the United States during the period 1973–1977, nontuberculousmycobacteria were isolated from more than 14% of specimens submitted<strong>to</strong> labora<strong>to</strong>ries on suspicion of tuberculosis (Thoen et al., 1979). More than 50% ofthe isolates corresponded <strong>to</strong> serotypes 1 <strong>and</strong> 2 of the M. avium complex; the rest primarilyconsisted of other serotypes from the same complex, <strong>and</strong> only 2.7% wereother species, such as M. fortuitum, M. paratuberculosis, M. kansasii, M. scrofulaceum,<strong>and</strong> M. xenopi.In São Paulo (Brazil), attempts at isolations from lesions in 28 cows <strong>and</strong> 62caseous lesions in slaughterhouse carcasses yielded 18 isolations of M. bovis <strong>and</strong> oneeach of M. tuberculosis, M. fortuitum, <strong>and</strong> M. kansasii (Correa <strong>and</strong> Correa, 1973).Although nontuberculous mycobacteria usually cause lesions only in lymphnodes, granulomas are sometimes found in other tissues.The principal problem presented by nontuberculous mycobacteria in cattle liesin the paraspecific sensitization for mammalian tuberculin, which causes confusionin diagnosis as well as the unnecessary slaughter of animals. The comparativetuberculin test (mammalian <strong>and</strong> avian) carried out in several countries shows thatsensitization <strong>to</strong> MAC is <strong>common</strong> in some countries <strong>and</strong> rare in others (Grangeet al., 1990).SWINE: In swine, MAC infection causes serious economic losses in many parts ofthe world due <strong>to</strong> confiscations of animals from slaughterhouses <strong>and</strong> lockers. In countriesthat have carried out successful programs <strong>to</strong> eradicate bovine tuberculosis, swineconfiscated for “tuberculosis” are primarily infected by MAC. Serotypes 1, 2, 4, 5,<strong>and</strong> 8 of this complex are the principal causes of mycobacterial infection in swine inthe United States (Songer et al., 1980). Serotype 8, in particular, has caused outbreakswith great losses for swine producers in several countries, including the UnitedStates, Japan, <strong>and</strong> South Africa. Lesions in these animals are usually restricted <strong>to</strong> cervical<strong>and</strong> mediastinal lymph gl<strong>and</strong>s, that is particularly near the digestive tract.Generalized lesions are usually due <strong>to</strong> M. bovis, but nontuberculous mycobacteriamay sometimes be responsible. In addition <strong>to</strong> the various MAC serotypes, other nontuberculousmycobacteria have also been isolated from swine, including M. kansasii


112 BACTERIOSES<strong>and</strong> M. fortuitum. Strains similar <strong>to</strong> M. fortuitum, but differing in several biochemicalcharacteristics, were isolated from swine with lymphadenitis; the name M.porcinum has been proposed for these strains (Tsukamura et al., 1983).MAC bacteria can sometimes be isolated from the apparently healthy lymphnodes of a large percentage of animals inspected in slaughterhouses (Brown <strong>and</strong>Neuman, 1979).In the US, any mycobacterial lesion is considered tuberculous for purposes ofinspecting pork. Economic losses due <strong>to</strong> tuberculosis were US$ 2.3 million in 1976,but fell 73% in 1988 (Dey <strong>and</strong> Parham, 1993).CATS AND DOGS: In cats, nodular lesions, with or without fistulation, are seen in thecutaneous <strong>and</strong> subcutaneous tissues, primarily on the venter. M. fortuitum is amongthe mycobacteria identified; on one occasion, M. xenopi was also found. This <strong>diseases</strong>hould be distinguished from “cat leprosy,” whose etiologic agent is M. lepraemurium<strong>and</strong> which is probably transmitted by rat bite. The cutaneous or subcutaneousnodules of “leprosy” can localize in any part of the body (White et al., 1983). Skininfections caused by nontuberculous mycobacteria also occur in dogs. Although dogsare resistant <strong>to</strong> MAC, 10 cases were confirmed in basset hounds; their susceptibilitymay be due <strong>to</strong> a genetic immunodeficiency (Carpenter et al., 1988).OTHER SPECIES: In addition <strong>to</strong> infections caused by the prevalent tuberculosismycobacteria (M. tuberculosis <strong>and</strong> M. bovis), infections caused by nontuberculousmycobacteria, such as various MAC serotypes, also occur in nonhuman primateskept in captivity. The infection is predominantly intestinal <strong>and</strong> manifests as diarrhea<strong>and</strong> emaciation. Lesions in these animals differ from those caused by M. tuberculosis<strong>and</strong> M. bovis in that tubercles do not form <strong>and</strong> necrosis <strong>and</strong> giant cells are absent.The lamina propria of the intestine is infiltrated by epithelioid cells (Thoen et al.,1981). In a cage of macaques (Macaca arc<strong>to</strong>ides), MAC infection was prevalentamong various <strong>diseases</strong> <strong>and</strong> caused the death of 44 of 54 animals over a period oftwo-<strong>and</strong>-a-half years. The lesions found upon au<strong>to</strong>psy indicated an enteric origin forthe disease process. His<strong>to</strong>pathological examination <strong>and</strong> clinical labora<strong>to</strong>ry examinationssuggested that the <strong>common</strong> basis of the <strong>diseases</strong> was an immunologic abnormality(Holmberg et al., 1985).Infection due <strong>to</strong> nontuberculous mycobacteria also occurs in other animal specieskept in captivity. In poikilotherms, the disease may be caused by various species ofmycobacteria, such as M. chelonae, M. marinum, M. fortuitum, <strong>and</strong> M. avium.An infection due <strong>to</strong> M. ulcerans was described in koalas (Phascolarc<strong>to</strong>s cinereus)on Raymond Isl<strong>and</strong> (Australia). The animals had ulcers on the flexor muscles oftheir extremities. This is the first confirmation of infection due <strong>to</strong> M. ulcerans in animalsother than man (Mitchell <strong>and</strong> Johnson, 1981).Disease among aquarium or aquiculture fish may be caused by several mycobacteria,particularly M. marinum <strong>and</strong> M. fortuitum. The clinical symp<strong>to</strong>ms are variable<strong>and</strong> may resemble other <strong>diseases</strong>, with emaciation, ascites, skin ulcerations, hemorrhages,exophthalmos, <strong>and</strong> skeletal deformities. Upon necropsy, grayish whitenecrotic foci are found in the viscera. Exposure <strong>to</strong> M. marinum in fish kept in aquariumsmay cause skin infections in man (Leibovitz, 1980; Martin, 1981).Unculturable mycobacteria that can be confused with M. leprae have been foundin several animal species, such as frogs in Bolivia (Pleurodema cinera <strong>and</strong> P. marmoratus)<strong>and</strong> water buffalo in Indonesia (Bubalus bubalis).


DISEASES CAUSED BY NONTUBERCULOUS MYCOBACTERIA 113In the province of Buenos Aires (Argentina), the lymph nodes of 67 apparentlynormal armadillos were cultured. Potentially pathogenic mycobacterial strains wereisolated from 22 (53.7%) of 41 hairy armadillos (Chae<strong>to</strong>phractus villosus) examined.These strains included M. intracellulare, M. fortuitum, <strong>and</strong> M. chelonae.Mycobacterial cultures were not obtained from 26 Dasypus hybridus armadillos(“mulitas”) (De Kan<strong>to</strong>r, 1978).To avoid errors, leprologists doing experimental work with armadillos must takein<strong>to</strong> account both identified mycobacteria from these animals as well as those insufficientlycharacterized <strong>to</strong> be identified (Resoagli et al.,1982).FOWL: Avian tuberculosis is due <strong>to</strong> M. avium serotypes 1, 2, <strong>and</strong> 3. Serotype 2 isthe most <strong>common</strong> in chickens, <strong>and</strong> serotype 1, in wild or captive birds in the US(Thoen et al., 1981). M. intracellulare is usually not pathogenic for fowl (Grange etal., 1990). The lesions are found mainly in the liver, spleen, intestine, <strong>and</strong> bone marrow,<strong>and</strong>, infrequently, in the lungs <strong>and</strong> kidneys. Avian tuberculosis is <strong>common</strong>; ithas a high incidence on farms where chickens have been kept for many years <strong>and</strong>the enclosures <strong>and</strong> grounds are contaminated. M. avium can survive in the soil forseveral years. In industrial establishments, the infection is rare because of the rapidreplacement of fowl, maintenance conditions, <strong>and</strong> hygienic measures.Turkeys can contract tuberculosis by living in association with infected chickens.Ducks <strong>and</strong> geese are not very susceptible <strong>to</strong> M. avium.The disease has been observed in several species of wild birds. It may affect anyspecies kept in zoos. Among birds kept as family pets, tuberculosis infections haveoccasionally been found in parrots, with M. tuberculosis as the etiologic agent causinginfections localized on the skin <strong>and</strong> in the natural orifices. This situation isexceptional among birds.Source of Infection <strong>and</strong> Mode of Transmission: Man <strong>and</strong> animals contract theinfection from environmental sources, such as water, soil, <strong>and</strong> dust. Human-<strong>to</strong>humantransmission has never been reliably demonstrated. M. fortuitum abounds innature <strong>and</strong> the ability of this mycobacteria, as well as M. chelonae, <strong>to</strong> multiply insoil has been confirmed experimentally. The natural hosts of serotypes 1, 2, <strong>and</strong> 3 ofM. avium are fowl, whose droppings help <strong>to</strong> contaminate the soil, which would bethe real reservoir. Other MAC serotypes have been isolated repeatedly from water.In one study (Gruft et al., 1981), MAIS complex mycobacteria were isolated from25% of 250 water samples collected along the eastern coast of the United States, primarilyfrom the warmer waters of the southeast coast. Similarly, isolations weremore abundant from estuarine samples than from river or sea water. During thisstudy, M. intracellulare was isolated from aerosols, which would explain the mechanismof transmission <strong>to</strong> man. Various MAC serotypes were also isolated from soil<strong>and</strong> house dust in research carried out in Australia <strong>and</strong> Japan. M. kansasii <strong>and</strong> M.xenopi were isolated from drinking water systems. The habitat of M. marinum iswater, <strong>and</strong> it has been isolated from snails, s<strong>and</strong>, <strong>and</strong> infected aquarium fish.Many nontuberculous mycobacteria are able <strong>to</strong> colonize the mucosa of thenasopharynx, bronchia, <strong>and</strong> intestines of immunocompetent people, who may experiencemycobacterial disease when their defenses are low. However, colonization isgenerally temporary in normal people, as demonstrated by PPD-A (avian) <strong>and</strong> PPD-B (Battey bacillus or M. intracellulare) tuberculin tests that become negative overtime. However, the more virulent MAC strains <strong>and</strong> serotypes that are able <strong>to</strong> estab-


114 BACTERIOSESlish themselves in normal subjects <strong>and</strong> immunodeficient individuals, such as AIDSpatients, now constitute an important pathogen.Nontuberculous mycobacteria are particularly abundant in soil contaminated byinfected animals, such as in pigsties, from where they may be carried <strong>to</strong> surfacewaters (Kazda, 1983).MAC <strong>and</strong> other mycobacteria can colonize drinking water. In a hospital in Bos<strong>to</strong>n(USA), MAC was cultured from 11 of 16 hot water faucets <strong>and</strong> shower heads, aswell as from 3 of 18 cold water faucets. Serotype 4 was predominant (du Moulin etal., 1988 cited in Grange et al., 1990).It is likely that the pulmonary disease in man is acquired through the respira<strong>to</strong>rysystem via aerosols. On the other h<strong>and</strong>, judging from the affected lymph nodes, lymphadenitisin man, cattle, <strong>and</strong> swine is possibly acquired through the intestine.Obviously, the mycobacteria that cause abscesses, cutaneous granulomas, <strong>and</strong> ulcerspenetrate through skin lesions.Tuberculosis in birds is transmitted by way of the intestine, through contaminatedfood, soil, <strong>and</strong> water.Role of Animals in the Epidemiology of the Disease: Mycobacteriosis is not azoonosis but rather a disease <strong>common</strong> <strong>to</strong> man <strong>and</strong> animals. Both acquire the infectionfrom environmental sources. Animals help <strong>to</strong> contaminate the environment, asin the case of birds <strong>and</strong> swine with MAC.Diagnosis: A reliable diagnosis can only be obtained through culture <strong>and</strong> identificationof the causal agent. The possibility of environmental contamination of theculture media should be kept in mind, as well as the fact that sputum, gastric wash,<strong>and</strong> saliva may contain nontuberculous mycobacteria without these causing disease.Repeated cultures with abundant growth of a potentially pathogenic Mycobacteriumspecies isolated from a patient with symp<strong>to</strong>ms consistent with the disease should beconsidered significant. The diagnosis is certain when nontuberculous mycobacteriaare isolated from surgical resection specimens. Differential diagnosis betweentubercular pulmonary infections (M. tuberculosis, M. bovis, <strong>and</strong> M. africanum) <strong>and</strong>nontuberculous mycobacterial infections is important, since M. avium-intracellulareis naturally resistant <strong>to</strong> anti-tuberculosis medications, while M. kansasii is sensitive<strong>to</strong> rifampicin <strong>and</strong> slightly resistant <strong>to</strong> the other medications (Wolinsky, 1979). Theother <strong>common</strong> forms of infection due <strong>to</strong> nontuberculous mycobacteria present fewerproblems in diagnosis.Infection in cattle <strong>and</strong> swine is generally diagnosed using lymph nodes obtainedin the slaughterhouse or lockers <strong>and</strong> sent <strong>to</strong> the labora<strong>to</strong>ry for culture.Clinical diagnosis of avian tuberculosis can be confirmed through au<strong>to</strong>psy <strong>and</strong>labora<strong>to</strong>ry techniques. The avian tuberculin test on the wattle is useful for diagnosingthe disease on farms. The agglutination test with whole blood is considered moreuseful in birds than the tuberculin test (Thoen <strong>and</strong> Karlson, 1991).The enzyme-linked immunosorbent assay (ELISA) has proven <strong>to</strong> be highly sensitivefor detecting antibodies <strong>to</strong> mycobacteria in swine, fowl, cattle, <strong>and</strong> other animals(Thoen et al., 1981).Control: Prevention of the pulmonary disease in man would consist of removingthe environmental sources of infection, which are difficult <strong>to</strong> recognize. Consequently,the recommended alternative is prevention <strong>and</strong> treatment of predisposing causes.


DISEASES CAUSED BY NONTUBERCULOUS MYCOBACTERIA 115Specific measures for preventing lymphadenitis in children are not available either. Onthe other h<strong>and</strong>, proper skin care, proper treatment of wounds, <strong>and</strong> avoidance of contaminatedswimming pools can prevent dermal <strong>and</strong> subcutaneous infections.The source of infection in swine affected by lymphadenitis has been determinedon several occasions, such as in cases described in Australia, the US, <strong>and</strong> Germany(Songer et al., 1980). When other materials were substituted for sawdust <strong>and</strong> shavingsused as bedding, the problem disappeared.The control of avian tuberculosis should focus primarily on farms. Given thelong-term survival of M. avium in the environment contaminated with the droppingsof tubercular fowl, the only remedy is <strong>to</strong> eliminate all existing birds on a farm <strong>and</strong>repopulate with healthy s<strong>to</strong>ck in an area not previously inhabited by fowl.Similar measures are needed <strong>to</strong> control mycobacteriosis in fish. Infected fishshould be destroyed <strong>and</strong> the aquarium disinfected. In addition, the introduction ofcontaminated fish or products should be avoided.BibliographyBarrera, L., I.N. De Kan<strong>to</strong>r. Nontuberculous mycobacteria <strong>and</strong> Mycobacterium bovis as acause of human disease in Argentina. Trop Geogr Med 39:222–227, 1987.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Blancarte, M., B. Campos, S. Serna Villanueva. Micobacterias atípicas en la RepúblicaMexicana. Salud Publica Mex 24:329–340, 1982.Brown, J., M.A. Neuman. Lesions of swine lymph nodes as a diagnostic test <strong>to</strong> determinemycobacterial infection. Appl Environ Microbiol 37:740–743, 1979.Brown, J., J.W. Tollison. Influence of pork consumption on human infection withMycobacterium avium-intracellulare. Appl Environ Microbiol 38:1144–1146, 1979.Carpenter, J.L., A.M. Myers, M.W. Conner, et al. Tuberculosis in five basset hounds. J AmVet Med Assoc 192:1563–1568, 1988.Correa, C.N., W.M. Correa. Micobacterias isoladas de bovinos e suinos em São Paulo,Brasil. Arq Inst Biol 40:205–208, 1973.Debrunner, M., M. Salfinger, O. Br<strong>and</strong>li, A. von Graevenitz. Epidemiology <strong>and</strong> clinical significanceof nontuberculous mycobacteria in patients negative for human immunodeficiencyvirus in Switzerl<strong>and</strong>. Clin Infect Dis 15:330–345, 1992.De Kan<strong>to</strong>r, I.N. Isolation of mycobacteria from two species of armadillos: Dasypushybridus (“mulita”) <strong>and</strong> Chae<strong>to</strong>phractus villosus (“peludo”). In: Pan American HealthOrganization. The Armadillo as an Experimental Model in Biomedical Research. Washing<strong>to</strong>n,D.C.: <strong>PAHO</strong>; 1978. (Scientific Publication 366).Dey, B.P., G.L. Parham. Incidence <strong>and</strong> economics of tuberculosis in swine slaughtered from1976 <strong>to</strong> 1988. J Am Vet Med Assoc 203:516–519, 1993.Di Lonardo, M., J. Benetucci, M. Beltrán, et al. La tuberculosis <strong>and</strong> la infección porVIH/SIDA en Argentina. Un resumen de información. Respiración 8:60–62, 1993.Di Lonardo, M., N.C. Isola, M. Ambroggi, G. Fulladosa, I.N. De Kan<strong>to</strong>r. Enfermedad producidapor micobacterias no tuberculosas en Buenos Aires, Argentina. Bol Oficina SanitPanam 95:134–141, 1983.Du Moulin, et al., 1988. Cited in: Grange, J.M., M.D. Yates, E. Bough<strong>to</strong>n. The avian tuberclebacillus <strong>and</strong> its relatives. J Appl Bacteriol 68:411–431, 1990.Grange, J.M., M.D. Yates, E. Bough<strong>to</strong>n. The avian tubercle bacillus <strong>and</strong> its relatives. J ApplBacteriol 68:411–431, 1990.


116 BACTERIOSESGruft, H., J.O. Falkinham III, B.C. Parker. Recent experience in the epidemiology of diseasecaused by atypical mycobacteria. Rev Infect Dis 3:990–996, 1981.Guthertz, L.S., B. Damsker, E.J. Bot<strong>to</strong>ne, et al. Mycobacterium avium <strong>and</strong> Mycobacteriumintracellulare infections in patients with <strong>and</strong> without AIDS. J Infect Dis 160:1037–1041, 1989.Havlik, J.A., Jr., C.R. Horsburgh, Jr., B. Metchock, et al. Disseminated Mycobacteriumavium complex infection: Clinical identification <strong>and</strong> epidemiologic trends. J Infect Dis165:577–580, 1992.Holmberg, C.A., R. Henrickson, R. Lenninger, et al. Immunologic abnormality in a groupof Macaca arc<strong>to</strong>ides with high mortality due <strong>to</strong> atypical mycobacterial <strong>and</strong> other diseaseprocesses. Am J Vet Res 46:1192–1196, 1985.Inderlied, C.B., C.A. Kemper, L.E. Bermúdez. The Mycobacterium avium complex. ClinMicrobiol Rev 6:266–310, 1993.Kazda, J. The principles of the ecology of mycobacteria. In: Ratledge C., J.L. Stanford, eds.Vol 2: The Biology of the Mycobacteria. London: Academic Press; 1983.Leibovitz, L. Fish tuberculosis (mycobacteriosis). J Am Vet Med Assoc 176:415, 1980.Martin, A.A. Mycobacteriosis: A brief review of a fish-transmitted zoonosis. In: Fowler,M.F., ed. Wildlife Diseases of the Pacific Basin <strong>and</strong> Other Countries. 4th InternationalConference of the Wildlife Diseases Association, Sydney, Australia, 1981.McFadden, J.J., P.D. Butcher, R. Chiodini, J. Hermon-Taylor. Crohn’s disease-isolatedmycobacteria are identical <strong>to</strong> Mycobacterium paratuberculosis, as determined by DNA probesthat distinguish between mycobacterial species. J Clin Microbiol 25:796–801, 1987.Mitchell, P., D. Johnson. The recovery of Mycobacterium ulcerans from koalas in eastGippsl<strong>and</strong>. In: Fowler, M.F., ed. Wildlife Diseases of the Pacific Basin <strong>and</strong> Other Countries.4th International Conference of the Wildlife Diseases Association, Sydney, Australia, 1981.Resoagli, E., A. Martínez, J.P. Resoagli, S.G. de Millán, M.I.O. de Rott, M. Ramírez.Micobacteriosis natural en armadillos, similar a la lepra humana. Gac Vet 44:674–676, 1982.Runyon, E.H. Anonymous mycobacteria in pulmonary disease. Med Clin North Am43:273–290, 1959.S<strong>and</strong>ers, W.E., Jr., E.A. Horowitz. Other Mycobacterium species. In: M<strong>and</strong>ell, G.L., R.G.Douglas, Jr., J.E. Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. NewYork: Churchill Livings<strong>to</strong>ne, Inc.; 1990.S<strong>and</strong>erson, J.D., M.T. Moss, M.L. Tizard, J. Hermon-Taylor. Mycobacterium paratuberculosisDNA in Crohn’s disease tissue. Gut 33:890–896, 1992.Schaefer, W.B. Incidence of the serotypes of Mycobacterium avium <strong>and</strong> atypical mycobacteriain human <strong>and</strong> animal <strong>diseases</strong>. Am Rev Respir Dis 97:18–23, 1968. Cited in: Wolinsky,E. Non tuberculous mycobacteria <strong>and</strong> associated <strong>diseases</strong>. Am Rev Resp Dis 119:107–159, 1979.Songer, J.G., E.J. Bicknell, C.O. Thoen. Epidemiological investigation of swine tuberculosisin Arizona. Can J Comp Med 44:115–120, 1980.Thoen, C.O., E.M. Himes, W.D. Richards, J.L. Jarnagin, R. Harring<strong>to</strong>n, Jr. Bovine tuberculosisin the United States <strong>and</strong> Puer<strong>to</strong> Rico: A labora<strong>to</strong>ry summary. Am J Vet Res 40:118–120, 1979.Thoen, C.O., A.G. Karlson. Tuberculosis. In: Calnek, B.W., H.J. Barnes, C.W. Beard, W.M.Reid, H.W. Yoder, Jr., eds. Diseases of Poultry. 9th ed. Ames: Iowa State UniversityPress; 1991.Thoen, C.O., A.G. Karlson, E.M. Himes. Mycobacterial infections in animals. Rev InfectDis 3:960–972, 1981.Thorel, M.F. Relationship between Mycobacterium avium, M. paratuberculosis <strong>and</strong>mycobacteria associated with Crohn’s disease. Ann Rech Vet 20:417–429, 1989.Thorel, M.F., M. Krichevsky, V.V. Levy-Frebault. Numerical taxonomy of mycobactindependentmycobacteria, emended description of Mycobacterium avium, <strong>and</strong> description ofMycobacterium avium subsp. avium subsp. nov., Mycobacterium avium subsp. paratubercu-


DISEASES IN MAN AND ANIMALS CAUSED BY NON-O1 VIBRIO CHOLERAE 117losis subsp. nov., <strong>and</strong> Mycobacterium avium subsp. silvaticum subsp. nov. Int J Syst Bacteriol40:254–260, 1990.Tsukamura, M., H. Nemo<strong>to</strong>, H. Yugi. Mycobacterium porcinum sp. nov. A porcinepathogen. Int J Syst Bacteriol 33:162–165, 1983.Wayne, L.G., G.P. Kubica. Family Mycobacteriaceae. In: Sneath, P.A., M.S. Mair,M.E. Sharpe. Vol 2: Bergey’s Manual of Systemic Bacteriology. Baltimore: Williams &Wilkins; 1986.White, S.D., P.J. Ihrke, A.A. Stannard, C. Cadmus, C. Griffin, S.A. Kruth, et al. Cutaneousatypical mycobacteriosis in cats. J Am Vet Med Assoc 182:1218–1222, 1983.Wolinsky, E. Nontuberculous mycobacteria <strong>and</strong> associated <strong>diseases</strong>. Am Rev Resp Dis119:107–159, 1979.Yakrus, M.A., R.C. Good. Geographic distribution, frequency, <strong>and</strong> specimen source ofMycobacterium avium complex serotypes isolated from patients with acquired immunodeficiencysyndrome. J Clin Microbiol 28:926–929, 1990.DISEASES IN MAN AND ANIMALS CAUSED BY NON-O1VIBRIO CHOLERAEICD-10 A00.0 cholera due <strong>to</strong> Vibrio cholerae O1, biovar choleraeEtiology: Vibrio cholerae, a slightly curved, comma-shaped, gram-negative,motile bacillus, 1.5 microns long by 0.4 microns in diameter. This species includesO1 V. cholerae, the etiologic agent of p<strong>and</strong>emic cholera, <strong>and</strong> non-O1 V. cholerae,which sometimes causes disease in man <strong>and</strong> animals.V. cholerae is serologically divided on the basis of its somatic O antigen. The etiologicalagents of typical, Asiatic, or epidemic cholera belong <strong>to</strong> serogroup O1. Allthe rest that do not agglutinate with the O1 antigen are non-O1 V. cholerae,formerlycalled nonagglutinable vibrios (NAGs).In March 1993, an epidemic strain of non-O1 V. cholerae was identified in SouthAsia <strong>and</strong> was designated as serogroup O139 (<strong>WHO</strong>, 1993). The first outbreakoccurred in November 1992 in Madras (India) <strong>and</strong> quickly assumed epidemic proportionsin India <strong>and</strong> Bangladesh, with thous<strong>and</strong>s of cases <strong>and</strong> high mortality (Daset al., 1993). Isolated strains of serogroup O139 produce a cholera <strong>to</strong>xin (CT) <strong>and</strong>hybridize with the CT’s DNA probe (Nair <strong>and</strong> Takeda, 1993). V. cholerae O139 containsa large number of gene copies of the <strong>to</strong>xin <strong>and</strong> is capable of producing it inlarge quantities so as <strong>to</strong> produce a severe pathogenic reaction (Das et al., 1993).Non-O1 Vibrio cholerae has biochemical <strong>and</strong> culturing properties that are verysimilar <strong>to</strong> those of the El Tor biotype of V. cholerae that is currently causing the seventhcholera p<strong>and</strong>emic, which began in Indonesia in 1958 <strong>and</strong> spread <strong>to</strong> a large par<strong>to</strong>f the Third World. Non-O1 V. cholerae does not agglutinate with a polyvalentserum against El Tor or against the Ogawa <strong>and</strong> Inaba subtypes. In addition, there isa great similarity between the O1 <strong>and</strong> non-O1 strains in numeric taxonomy, isoenzymeanalysis, <strong>and</strong> DNA:DNA hybridization analysis (Benenson, 1991).


118 BACTERIOSESThere are various schemes that classify non-O1 V. cholerae in serovars (orserotypes). One of them (currently used in the US) is the Smith scheme (Smith,1977), which distinguishes more than 70 serovars. Serotyping is limited <strong>to</strong> referencelabora<strong>to</strong>ries for epidemiologic studies.Geographic Distribution: Worldwide. The presence of non-O1 V. cholerae hasbeen confirmed on all inhabited continents, either in the environment (particularlyin bodies of water), or in man <strong>and</strong> animals. In Asia, serogroup O139 has spread fromBangladesh <strong>and</strong> India <strong>to</strong> China, Malaysia, Nepal, Pakistan, <strong>and</strong> Thail<strong>and</strong>, <strong>and</strong> mayspread further. The first case introduced in<strong>to</strong> the US was a California resident whohad traveled <strong>to</strong> India. There was also a case in the UK.Occurrence in Man: In man, it appears as sporadic cases or small outbreaks. Inareas where cholera is endemic, patients have frequently suffered a disease similar<strong>to</strong> cholera, but caused by non-O1 V. cholerae. In India <strong>and</strong> Pakistan, nonagglutinablevibrios were isolated (i.e., non-O1 V. cholerae) from a small percentage ofpatients with choleriform symp<strong>to</strong>ms. In 1968, an outbreak attributed <strong>to</strong> this agen<strong>to</strong>ccurred in Sudan <strong>and</strong> caused gastroenteritis in 544 people, 31 of whom died(Kamal, 1971). In the former Czechoslovakia, an outbreak of gastroenteritis affected56 young people at a training center. NAGs were isolated from 42 of the 56, but notfrom 100 controls. The disease was attributed <strong>to</strong> this etiologic agent <strong>and</strong> the vehicleof infection was thought <strong>to</strong> be pota<strong>to</strong>es (possibly contaminated after cooking) thatthe patients ate. The disease was mild <strong>and</strong> short lived (Aldová et al., 1968). Therewas also an outbreak on a flight from London <strong>to</strong> Australia that was attributed <strong>to</strong> anasparagus <strong>and</strong> egg salad (Dakin et al., 1974). Sporadic cases are more <strong>common</strong> <strong>and</strong>have occurred in several countries.The appearance of serogroup O139 completely changed the scenario. Thisserogroup is not distinguished from serogroup O1 as an epidemic agent of cholera.The epidemic it is causing has affected tens of thous<strong>and</strong>s of people, with approximatelya 5% mortality rate (<strong>WHO</strong>, 1993). The fear is that this new agent has thepotential <strong>to</strong> cause a p<strong>and</strong>emic. The epidemic wave has already moved from India <strong>to</strong>Thail<strong>and</strong>, Bangladesh, <strong>and</strong> other countries.Occurrence in Animals: Non-O1 V. cholerae has been isolated from manydomestic <strong>and</strong> wild mammalian species, as well as from birds. In India, 14% of morethan 500 dogs harbored “noncholeric vibrios” in their intestines. In the same geographicarea, the same vibrios were found in ravens (Sack, 1973). In another area ofIndia, far from the endemic cholera area, 195 domestic animals were examined(goats, cows, dogs, <strong>and</strong> birds) in a search for an animal reservoir for V. cholerae.Fifty-four strains were isolated, 8 of which were O1 V. cholerae <strong>and</strong> 46 of whichwere non-O1. Serotype O1 was found only during the months when cholera washighly prevalent in the population, whereas the other serotype was found throughoutthe year (Sanyal et al., 1974).The Disease in Man: It appears in two forms: intestinal, which is prevalent, <strong>and</strong>extraintestinal.Gastroenteritis caused by non-O1 V. cholerae is usually of short duration <strong>and</strong> thesymp<strong>to</strong>ms are mild <strong>to</strong> moderate. The disease is only occasionally severe, as occursin epidemic cholera (Morris, 1990). The clinical picture is usually variable. In agroup of 14 patients in the US, 100% had diarrhea (25% of the patients had bloody


DISEASES IN MAN AND ANIMALS CAUSED BY NON-O1 VIBRIO CHOLERAE 119diarrhea), 93% had abdominal pain, 71% had fever, <strong>and</strong> 21% had nausea <strong>and</strong> vomiting.Eight of the 14 patients required hospitalization (Morris, 1990). A severe diseasewas diagnosed in two young <strong>to</strong>urists who returned <strong>to</strong> Canada from theDominican Republic (Girouard et al., 1992).Of three strains given <strong>to</strong> volunteers, only one reproduced the diarrheal diseasewith s<strong>to</strong>ol volumes of 140 ml <strong>to</strong> 5,397 ml (Morris et al., 1990).In contrast with epidemic V. cholerae,which is exclusively intestinal, non-O1 wasisolated from different localizations, such as blood (20,8%), wounds (approximately7%), the respira<strong>to</strong>ry tract (5%), the ears (11.9%), <strong>and</strong> others (cystitis, cellulitis,peri<strong>to</strong>nitis).Septicemia caused by non-O1 V. cholerae occurs primarily in immunodeficientindividuals (chronic hepa<strong>to</strong>pathy, malignant hema<strong>to</strong>logical <strong>diseases</strong>, transplants),with a fatal outcome in more than 60% of cases. In other localizations, non-O1 isoften found with other pathogens, <strong>and</strong> thus it is difficult <strong>to</strong> discern its true role(Morris, 1990). A case of spontaneous peri<strong>to</strong>nitis <strong>and</strong> sepsis caused by non-O1 vibriowas described in Argentina. The underlying disease was hepatitis B <strong>and</strong> non-O1Vibrio cholerae was isolated through blood culture as well as from ascitic fluid in apure state (Soloaga et al., 1991). In addition, serotype O139 was isolated from theblood of a hepatic patient in India, something that does not occur with patients sufferingfrom cholera caused by O1 V. cholerae.The disease caused by serogroup O139 is not distinguished from that caused byO1. Infection due <strong>to</strong> O1 Vibrio cholerae apparently does not confer cross immunityagainst O139, as the latter occurs in areas where people of all ages should have somelevel of immunity <strong>to</strong> cholera.In severe cases, dehydration must be treated through fluid <strong>and</strong> electrolyte replacement.In addition, patients should be treated with tetracycline.Treatment for patients infected by O139 is the same as for patients infected by O1V. cholerae: rehydration <strong>and</strong>, in severe cases, administration of tetracycline.The Disease in Animals: There are few records of the disease in animals; mostspecies seem <strong>to</strong> be asymp<strong>to</strong>matic carriers.In western Colorado (USA), there was an outbreak of the disease in which 7 ofapproximately 100 American bison (Bison bison) died in about three days. The sickbison were depressed <strong>and</strong> separated themselves from the rest of the herd. The principalsymp<strong>to</strong>ms were diarrhea, vomiting, serous nasal discharge, weepy eyes, <strong>and</strong>conjunctival congestion. Upon necropsy, lesions were found only in the digestivetract. Non-O1 V. cholerae was isolated from the abomasum, duodenum, <strong>and</strong> colonof one animal <strong>and</strong> from an intestinal swab from another animal. The agent had beenisolated previously in the same region from a colt <strong>and</strong> a sheep (Rhodes et al., 1985).Rhodes et al. (1986) studied several bodies of water, from freshwater <strong>to</strong> salt water(17 mmol of sodium/L). In western Colorado, 16 different serovars of non-O1 V.cholerae were found.In Argentina, an outbreak occurred in young bulls, affecting 20% of 800 animals,with 50% mortality. The main symp<strong>to</strong>m was diarrhea with dark green feces <strong>and</strong>weight loss. Upon necropsy, hypertrophy of the mesenteric lymph node chain wasfound. A bacterium with the characteristics of non-O1 V. cholerae was isolated fromthe lymph nodes (Fain Binda et al., 1986).In addition <strong>to</strong> these outbreaks, sporadic cases have been recorded in several countries.


120 BACTERIOSESSource of Infection <strong>and</strong> Mode of Transmission: Non-O1 V. cholerae is a naturalinhabitant of the surface waters of estuaries, rivers, streams, lakes, irrigationchannels, <strong>and</strong> the sea. It has also been isolated from wastewater from an Argentinecity (Corrales et al., 1989). Thus, water constitutes the principal reservoir of this etiologicagent.Non-O1 V. cholerae has been isolated from many animal species in different partsof the world. However, their role as reservoirs is still in dispute (Morris, 1990).Man can be a carrier of the agent <strong>and</strong> the source of infection for others. In a studyconducted on Iranian pilgrims returning from Mecca, several of their contactsacquired the infection <strong>and</strong> had diarrhea (Zafarí et al., 1973). The source of infectionis different in each country. In the US, the main source of infection is raw oysters.Of 790 samples of fresh oysters, 14% contained non-O1 V. cholerae. The number ofisolations was higher in the summer months, when there are more vibrios in thewater (Twedt et al., 1981). As expected, the highest number of human cases has alsooccurred in summer <strong>and</strong> autumn. A variety of contaminated foods have been implicatedin other countries (see the section on occurrence in man). Surface water adjacent<strong>to</strong> a cistern was possibly the vehicle of infection for an outbreak in Sudan in1968 (Kamal, 1971). In a refugee camp in Thail<strong>and</strong>, 16% of drinking water sampleswere contaminated by non-O1 V. cholerae (Taylor et al., 1988).A case of cystitis occurred in a woman after she swam in Chesapeake Bay (USA).Ear <strong>and</strong> wound infections have almost always been caused by exposure <strong>to</strong> seawater.It is more difficult <strong>to</strong> establish the source of infection in septicemias. Some are associatedwith diarrhea, which would indicate infection via the oral route. Shellfishhave been suspected in several cases.Only a minority of strains of non-O1 V. cholerae are pathogenic. At present, it canbe stated that strains isolated from patients are more virulent than environmentalstrains. Strains are differentiated based on hemolysins, their ability <strong>to</strong> colonize theintestine (adherence fac<strong>to</strong>r), <strong>and</strong> the production of a <strong>to</strong>xin similar <strong>to</strong> cholera <strong>to</strong>xin,Shiga-like <strong>to</strong>xin, <strong>and</strong> a thermostable entero<strong>to</strong>xin similar <strong>to</strong> that produced by entero<strong>to</strong>xigenicEscherichia coli. In India <strong>and</strong> Bangladesh, non-O1 strains that producecholera <strong>to</strong>xin were isolated, but this happens less frequently in other countries. InThail<strong>and</strong>, only 2% of 237 environmental non-O1 strains <strong>and</strong> none of 44 strains isolatedfrom clinical cases carried gene sequences homologous with the cholera <strong>to</strong>xingene. In summary, strains of V. cholerae vary greatly in terms of the fac<strong>to</strong>rs thatcould determine virulence <strong>and</strong> no single characteristic has been identified that couldbe used <strong>to</strong> differentiate pathogenic strains from avirulent strains (Morris, 1990).Role of Animals in the Epidemiology of the Disease: Although the agent hasbeen isolated from many animal species <strong>and</strong> many researchers consider such animalsreservoirs or possible sources of infection for man (Sack, 1973; Sanyal et al.,1974), their actual role is questionable.Diagnosis: Culture, isolation, <strong>and</strong> characterization of the microorganism is theonly irrefutable method for diagnosing the disease. Alkaline pep<strong>to</strong>ne water (APW)<strong>and</strong> Monsur broth with tellurite <strong>and</strong> bile salts are useful enrichment media. The recommendedselective medium is thiosulfate citrate bile salts sucrose agar (TCBS)(Corrales et al., 1989).The corresponding antiserum should be used for specific diagnosis of the O139strain (<strong>WHO</strong>, 1993).


DISEASES IN MAN AND ANIMALS CAUSED BY NON-O1 VIBRIO CHOLERAE 121Control <strong>and</strong> Prevention: The few recommendations that can be made are not <strong>to</strong>eat raw or inadequately cooked shellfish <strong>and</strong> other seafood, <strong>and</strong> <strong>to</strong> drink onlypotable water.To prevent infection by serogroup O139, the same recommendations as for classiccholera (serogroup O1) apply.BibliographyAldová, E., K. Laznickova, E. Stepankova, J. Lietava. Isolation of nonagglutinable vibriosfrom an enteritis outbreak in Czechoslovakia. J Infect Dis 118:25–31, 1968. Cited in: Morris,J.G., Jr. Non-O group 1 Vibrio cholerae: A look at the epidemiology of an occasionalpathogen. Epidemiol Rev 12:179–191, 1990.Benenson, A.S. Cholera. In: Evans, A.S., P.S. Brachman, eds. Bacterial Infections ofHumans. 2nd ed. New York: Plenum Medical Book Co.; 1991.Corrales, M.T., G.B. Fronchkowsky, T. Eiguer. Aislamien<strong>to</strong> en Argentina de Vibrio choleraeno O1 en líquidos cloacales. Rev Argent Microbiol 21:71–77, 1989.Dakin, W.P., D.J. Howell, R.G. Sut<strong>to</strong>n, et al. Gastroenteritis due <strong>to</strong> non-agglutinable(non-cholera) vibrios. Med J Aust 2:487–490, 1974. Cited in: Morris, J.G., Jr. Non-O group 1Vibrio cholerae: A look at the epidemiology of an occasional pathogen. Epidemiol Rev12:179–191, 1990.Das, B., R.K. Ghosh, C. Sharma, et al. T<strong>and</strong>em repeats of cholera <strong>to</strong>xin gene in Vibriocholerae O139. Lancet 342(8880):1173–1174, 1993.Fain Binda, J.C., E.R. Comba, H. Sánchez, et al. Primer aislamien<strong>to</strong> de Vibrio cholerae noO1 en la Argentina de una enteritis bovina. Rev Med Vet 67:203–207, 1986.Girouard, Y., C. Gaudreau, G. Frechette, M. Lorange-Rodriques. Non-O1 Vibrio choleraeenterocolitis in Quebec <strong>to</strong>urists returning from the Dominican Republic. Can Commun DisRep 18:105–107, 1992.Kamal, A.M. Outbreak of gastroenteritis by nonagglutinable (NAG) vibrios in the Republicof the Sudan. J Egypt Public Health Assoc 46:125–173, 1971. Cited in: Benenson, A.S.Cholera. In: Evans, A.S., P.S. Brachman, eds. Bacterial Infections of Humans. 2nd ed. NewYork: Plenum Medical Book Co.; 1991.Kaper, J., H. Lockman, R.R. Colwell, S.W. Joseph. Ecology, serology, <strong>and</strong> entero<strong>to</strong>xin productionof Vibrio cholerae in Chesapeake Bay. Appl Environ Microbiol 37:91–103, 1979.Morris, J.G., Jr. Non-O group 1 Vibrio cholerae: A look at the epidemiology of an occasionalpathogen. Epidemiol Rev 12:179–191, 1990.Morris, J.G., Jr., T. Takeda, B.D. Tall, et al. Experimental non-O group 1 Vibrio choleraegastroenteritis in humans. J Clin Invest 85:697–705, 1990.Nair, G.B., Y. Takeda. Vibrio cholerae in disguise: A disturbing entity. Wld J MicrobiolBiotech 9:399–400, 1993.Rhodes, J.B., D. Schweitzer, J.E. Ogg. Isolation of non-O1 Vibrio cholerae associated withenteric disease of herbivores in western Colorado. J Clin Microbiol 22:572–575, 1985.Rhodes, J.B., H.L. Smith, Jr., J.E. Ogg. Isolation of non-O1 Vibrio cholerae serovars fromsurface waters in western Colorado. Appl Environ Microbiol 51:1216–1219, 1986.Sack, R.B. A search for canine carriers of Vibrio. J Infect Dis 127:709–712, 1973.Sanyal, S.C., S.J. Singh, I.C. Tiwari, et al. Role of household animals in maintenance ofcholera infection in a community. J Infect Dis 130:575–579, 1974.Smith, H.L. Serotyping of non-cholera vibrios. J Clin Microbiol 30:279–282, 1977.Soloaga, R., G. Martínez, A. Cattani, et al. Peri<strong>to</strong>nitis bacteriana espontánea y sepsis porVibrio cholerae no O1. Infect Microbiol Clin 3:58–62, 1991.Taylor, D.N., P. Echeverría, C. Pitarangsi, et al. Application of DNA hybridization techniquesin the assessment of diarrheal disease among refugees in Thail<strong>and</strong>. Am J Epidemiol


122 BACTERIOSES127:179–187, 1988. Cited in: Morris, J.G., Jr. Non-O group 1 Vibrio cholerae:A look at theepidemiology of an occasional pathogen. Epidemiol Rev 12:179–191, 1990.Twedt, R.M., J.M. Madden, J.M. Hunt, et al. Characterization of Vibrio cholerae isolatedfrom oysters. Appl Environ Microbiol 41:1475–1478, 1981.World Health Organization (<strong>WHO</strong>). Epidemic diarrhoea due <strong>to</strong> Vibrio cholerae non-O1.Wkly Epidemiol Rec 68(20):141–142, 1993.Zafarí, Y., S. Rahmanzadeh, A.Z. Zarifi, N. Fakhar. Diarrhoea caused by non-agglutinableVibrio cholerae (non-cholera Vibrio). Lancet 2:429–430, 1973. Cited in: Morris, J.G., Jr. Non-O group 1 Vibrio cholerae:A look at the epidemiology of an occasional pathogen. EpidemiolRev 12:179–191, 1990.


ENTEROCOLITIC YERSINIOSISICD-10 A04.6 enteritis due <strong>to</strong> Yersinia enterocoliticaEtiology: Yersinia enterocolitica is a gram-negative coccobacillus that is motileat 25°C <strong>and</strong> belongs <strong>to</strong> the family Enterobacteriaceae. This species includes a veryheterogeneous group of bacteria that differ greatly in their biochemical properties.Currently, the biochemically atypical strains have been classified as seven differentadditional species: Y. aldovae, Y. bercovieri, Y. frederiksenii, Y. intermedia, Y. mollaretii,Y. kristensenii, <strong>and</strong> Y. rohdei. These are generally environmental species, canbe confused with Y. enterocolitica, <strong>and</strong> at times cause some extraintestinal infections(Farmer <strong>and</strong> Kelly, 1991). The suggestion has been made <strong>to</strong> subdivide thespecies Y. enterocolitica in<strong>to</strong> biotypes <strong>and</strong> serotypes. Biotyping is based on biochemicalcharacteristics, while serotyping is based on the O antigen. The species hasbeen subdivided in<strong>to</strong> more than 50 serotypes, but only some have proven <strong>to</strong> be pathogenicfor man or animals. More recently, the use of ribotyping was suggested forserotype O:3, permitting the differentiation of four clones. Most O:3 isolates belong<strong>to</strong> clones I <strong>and</strong> II. These same ribotypes were isolated in Japan. Ribotype I wasisolated in Canada <strong>and</strong> ribotypes II <strong>and</strong> IV were isolated in Belgium (Blumberget al., 1991).A 40- <strong>to</strong> 50-megadal<strong>to</strong>n plasmid is apparently responsible for the virulence of Y.enterocolitica. Strains with this plasmid are characterized by au<strong>to</strong>agglutination, calciumdependence (antigens V <strong>and</strong> W cause dependence on calcium for growth at37°C), <strong>and</strong> absorption of Congo red. There are also strains that do not contain theplasmid; these are pathogenic <strong>and</strong> generally negative <strong>to</strong> pyrazinamide, salicin, <strong>and</strong>aesculin (Riley <strong>and</strong> Toma, 1989). Several researchers have found inconsistenciesbetween virulence markers <strong>and</strong> disease. A study conducted in Santiago (Chile) on acohort of children up <strong>to</strong> 4 years of age isolated Y. enterocolitica from the feces of1.1% of children with diarrhea <strong>and</strong> from 0.2% of the controls. In a subgroup of thiscohort, 6% of the children with weekly fecal cultures were bacteriologically posi-


ENTEROCOLITIC YERSINIOSIS 123tive without presenting clinical symp<strong>to</strong>ms. The isolates of Y. enterocolitica from theasymp<strong>to</strong>matic children were serotype O:3, but did not have the virulence propertiesattributed <strong>to</strong> virulent strains (Morris et al., 1991).Geographic Distribution: Worldwide. The agent has been isolated from animals,man, food, <strong>and</strong> water. The human disease has been confirmed on five continents <strong>and</strong>in more than 30 countries (Swaminathan et al., 1982). There are geographic differencesin the distribution of serotypes. Serotypes 3, 5, 9, <strong>and</strong> 27 are found in Europe<strong>and</strong> many countries on other continents with temperate or cold climates. Serotypes8, 13, 18, 20, <strong>and</strong> 21 appear primarily in the US. Serotype 8 has caused several epidemicoutbreaks (Carniel <strong>and</strong> Mollaret, 1990). Outside the Americas, serotype 8 hasbeen isolated from the fecal matter of a healthy dog <strong>and</strong> a piglet in Nigeria. None ofthese strains had virulence markers (Trimnell <strong>and</strong> Adesiyun, 1988).The serotype pattern is changing in the US. In New York City <strong>and</strong> New York State,serotype 3 appears most frequently; this is also true in California. From 1972 <strong>to</strong>1979, only two isolates of O:3 were confirmed in California, but the frequency ofthis serotype began <strong>to</strong> increase such that from 1986 <strong>to</strong> 1988 it was part of 41% ofall isolates of Y. enterocolitica (Bissett et al., 1990). This trend seems <strong>to</strong> be spreadingas serotype O:3 in children has emerged in Atlanta, Georgia <strong>and</strong> in other UScities (Lee et al., 1991).Occurrence in Man: There are marked differences in disease incidence betweendifferent regions <strong>and</strong> even between neighboring countries. The highest incidencerates are observed in Sc<strong>and</strong>inavia, Belgium, several eastern European countries,Japan, South Africa, <strong>and</strong> Canada. The disease is less <strong>common</strong> in the United States,Great Britain, <strong>and</strong> France. In Belgium, the agent was isolated from 3,167 patientsbetween 1963 <strong>and</strong> 1978, with isolations increasing since then. Of the strains isolated,84% belonged <strong>to</strong> serotype 3, but isolations of serotype 9 have risen since then(de Groote et al., 1982). In Canada from 1966 <strong>to</strong> 1977, 1,000 isolations (serotype 3)were made from human patients, while in the US from 1973 <strong>to</strong> 1976, 68 casesoccurred, with serotype 8 predominating. Approximately 1% <strong>to</strong> 3% of acute enteritiscases in Sweden, the former West Germany, Belgium, <strong>and</strong> Canada are caused byY. enterocolitica (<strong>WHO</strong> Scientific Working Group, 1980). The lack of labora<strong>to</strong>ryfacilities hinders knowledge of disease incidence in developing countries. In tropicalareas, Y. enterocolitica seems <strong>to</strong> be a minor cause of diarrhea (Mata <strong>and</strong>Simhon, 1982).Most cases are sporadic or appear as small, familial outbreaks, but several epidemicshave also been described. Three of these outbreaks occurred in Japan in 1972<strong>and</strong> affected children <strong>and</strong> adolescents, with 189 cases in one epidemic, 198 inanother, <strong>and</strong> 544 in the third. The source of infection could not be determined. In1976, an outbreak in the state of New York affected 218 school children. The sourceof the infection was thought <strong>to</strong> be chocolate milk (possibly made with contaminatedchocolate syrup). An outbreak in 1982 in the US affected three states (Tennessee,Arkansas, <strong>and</strong> Mississippi), <strong>and</strong> caused 172 patients <strong>to</strong> be hospitalized. Serotypes 13<strong>and</strong> 18 of Y. enterocolitica, which are rare in the US, were isolated from thesepatients. Statistical association indicated milk from a single processing plant as thesource of infection (Tacket et al., 1984). An outbreak was reported in 1973 inFinl<strong>and</strong> that affected 94 conscripts (Lindholm <strong>and</strong> Visakorpi, 1991). A study conductedin a hospital in the Basque country of Spain on 51 cases of yersiniosis


124 BACTERIOSESrecorded during the period 1984–1989 found that most of the patients were urban,62% were male, their average age was 16–19 years, <strong>and</strong> the hospital stay was 6 <strong>to</strong>12 days for adults <strong>and</strong> less for children (Franco-Vicario et al., 1991). However, thisscenario varies from country <strong>to</strong> country. In many industrialized countries, Y. enterocoliticais one of the principal causes of gastroenteritis in children <strong>and</strong> sometimes issecond <strong>to</strong> Salmonella in isolates taken from the pediatric population (Cover <strong>and</strong>Aber, 1989). In late 1989 <strong>and</strong> early 1990, an outbreak occurred in Atlanta, Georgia(USA) among black children. Y. enterocolitica serotype 3 was isolated from 38(0.78%) of 4,841 fecal samples from seven hospitals in different American cities.Twenty of the 38 children had eaten pig intestines (“chitterlings”), which were probablyundercooked. Other intestinal pathogens isolated were Shigella (1.01%),Campylobacter (1.24%), <strong>and</strong> Salmonella (2.02%) (Lee et al., 1991). An outbreakaffecting 80 children was recorded in Rumania (Constantiniu et al., 1992).Most cases occur in fall <strong>and</strong> winter in Europe <strong>and</strong> from December <strong>to</strong> May inSouth Africa.Occurrence in Animals: Y. enterocolitica has been isolated from many domestic<strong>and</strong> wild mammals, as well as from some birds <strong>and</strong> cold-blooded animals. Theserotypes isolated from most animal species differ from those in man. Importantexceptions are swine, dogs, <strong>and</strong> cats, from which serotypes 3 <strong>and</strong> 9, the most prevalentcauses of human infection in many countries, have been isolated. In addition,serotype 5 was found in swine <strong>and</strong> is <strong>common</strong> in people in Japan (Hurvell, 1981).In some countries, the rate of isolations from animals is very high. In Belgium,serotypes that affect man were isolated from 62.5% of pork <strong>to</strong>ngues collected frombutchers (de Groote et al., 1982). Studies done in Belgium <strong>and</strong> Denmark revealedthat 3% <strong>to</strong> 5% of swine carry the agent in their intestines.The Disease in Man: Y. enterocolitica is mainly a human pathogen that usuallyaffects children. The predominant symp<strong>to</strong>m in small children is an acute enteritiswith watery diarrhea lasting 3 <strong>to</strong> 14 days; blood is present in the s<strong>to</strong>ol in 5% of cases.In older children <strong>and</strong> adolescents, pseudoappendicitis syndrome predominates, withpain in the right iliac fossa, fever, moderate leukocy<strong>to</strong>sis, <strong>and</strong> a high rate of erythrosedimentation.The syndrome’s great similarity <strong>to</strong> acute appendicitis has frequentlyled <strong>to</strong> surgery. In adults, especially in those over 40 years of age, an erythemanodosum may develop one <strong>to</strong> two weeks after enteritis. The prognosis is favorable foralmost all those affected, 80% of whom are women. Reactive arthritis of one or morejoints is a more serious complication. About 100 cases of septicemia have beendescribed, mainly in Europe. Other complications may be present, but are much rarer.Of 1,700 patients with Y. enterocolitica infection in Belgium, 86% had gastroenteritis,nearly 10% had the pseudoappendicitis syndrome, <strong>and</strong> less than 1% had septicemia<strong>and</strong> hepatic abscesses (Swaminathan et al., 1982).An epidemic with 172 cases occurred in the United States in 1982 <strong>and</strong> was attributed<strong>to</strong> pasteurized milk: 86% had enteritis <strong>and</strong> 14% had extraintestinal infectionslocalized in the throat, blood, urinary tract, peri<strong>to</strong>neum, central nervous system, <strong>and</strong>wounds. Extraintestinal infections were more <strong>common</strong> in adults. In patients withenteritis (mostly children), the disease caused fever (92.7%), abdominal pain(86.3%), diarrhea (82.7%), vomiting (41.4%), sore throat (22.2%), cutaneous eruptions(22.2%), bloody s<strong>to</strong>ol (19.7%), <strong>and</strong> joint pain (15.1%). The last symp<strong>to</strong>m wasseen only in patients 3 years of age or older (Tacket et al., 1984).


ENTEROCOLITIC YERSINIOSIS 125Although extraintestinal complications are rare, they can be fatal (mortality isestimated at 34% <strong>to</strong> 50%) (Marasco et al., 1993). Complications, such as hepatic orsplenic abscesses, occur in adults <strong>and</strong> generally in immunodeficient patients.Mortality is very high in septicemia caused by transfusion of red blood cells contaminatedby Y. enterocolitica. Of 35 cases counted, 23 were fatal. Fever <strong>and</strong>hypotension are the principal symp<strong>to</strong>ms <strong>and</strong> appear in less than one hour (see thesection on source of infection <strong>and</strong> mode of transmission).In Norway during the period 1974–1983, 458 cases of yersiniosis were diagnosed<strong>and</strong> patients were followed up for 10 years. Upon admission <strong>to</strong> the hospital, 184patients had abdominal pain, 200 had diarrhea, 45 experienced vomiting, <strong>and</strong> 36experienced weight loss. Mesenteric lymphadenitis or ileitis was found in 43 of 56who underwent laparo<strong>to</strong>my. Four <strong>to</strong> 14 years after discharge, 38 were readmittedwith abdominal pain, <strong>and</strong> 28 with diarrhea. High mortality was confirmed in 16 of22 patients who suffered from chronic hepatitis as a result of the infection (Saebo<strong>and</strong> Lassen, 1992a <strong>and</strong> 1992b).Treatment may be useful in the case of gastrointestinal symp<strong>to</strong>ms <strong>and</strong> is highlyrecommended for septicemia <strong>and</strong> complications from the disease (Benenson, 1992).Y. enterocolitica is susceptible <strong>to</strong> <strong>common</strong>ly used antimicrobials, except for ampicillin<strong>and</strong> cephalothin. There are indications that there is no good correlationbetween in vitro assays <strong>and</strong> clinical efficacy (Lee et al., 1991). Aminoglycosides arethe antibiotics recommended most often in cases of septicemia. Other indicatedantimicrobials are cotrimoxazole <strong>and</strong> ciprofloxacin.The Disease in Animals: In the 1960s, several epizootics in chinchillas occurredin Europe, the United States, <strong>and</strong> Mexico, with many cases of septicemia <strong>and</strong> highmortality. These outbreaks were originally attributed <strong>to</strong> Y. pseudotuberculosis, butthe agent was later determined <strong>to</strong> be Y. enterocolitica serotype 1 (biotype 3), whichhad never been isolated from man. The principal clinical symp<strong>to</strong>ms consisted ofsialorrhea, diarrhea, <strong>and</strong> weight loss. In the same period, cases of septicemia weredescribed in hares, from which serotype 2 (biotype 5) was isolated; this serotypealso does not affect man. Y. enterocolitica has been isolated from several species ofwild animals, in some of which intestinal lesions or hepatic abscesses were found.In the former Czechoslovakia <strong>and</strong> the Sc<strong>and</strong>inavian countries, Y. enterocolitica wasisolated from 3% <strong>to</strong> 26% of wild rodents, but necropsy of these animals revealed nolesions. Similar results were obtained in southern Chile, where the agent was isolatedfrom 4% of 305 rodents of different species <strong>and</strong> from different habitats(Zamora et al., 1979). Serotypes isolated from rodents are generally not those pathogenicfor man. Among wild animals in New York State, serotype O:8 has been isolatedfrom a gray fox (Urocyon cinereoargenteus) <strong>and</strong> from a porcupine (Erethizondorsatum); serotype O:3 has been isolated from another gray fox. Both serotypes arepathogenic for man (Shayegani et al., 1986).Studies carried out on swine, dogs, <strong>and</strong> cats are of particular interest, since theseanimals harbor serotypes that infect man. The agent has been isolated from clinicallyhealthy swine <strong>and</strong> from animals destined for human consumption. In onestudy, a much higher rate of isolations was obtained from swine with diarrhea thanfrom apparently healthy animals. In another study, however, the agent was isolatedfrom 17% of healthy swine <strong>and</strong> from 5.4% of swine tested because of various symp<strong>to</strong>ms(Hurvell, 1981). Y. enterocolitica has been isolated from swine during out-


126 BACTERIOSESbreaks of diarrhea, with no other pathogen detected. Blood or mucus do not generallyappear in the feces, but may be found in the s<strong>to</strong>ol of some animals. Diarrhea isaccompanied by a mild fever (Taylor, 1992). Swine that are carriers of Y. enterocoliticaserotypes that infect man have been noted primarily in countries where theincidence of human disease is higher, such as in the Sc<strong>and</strong>inavian countries,Belgium, Canada, <strong>and</strong> Japan. The rate of isolation from swine varies from one herd<strong>to</strong> another <strong>and</strong> depends on the level of contamination in each establishment. On onefarm the agent may be isolated only sporadically <strong>and</strong> at a low rate, while on another,isolations may be continuous <strong>and</strong> reach 100% of the groups examined (Fukushimaet al., 1983).Y. enterocolitica has been isolated from young sheep with enterocolitis in NewZeal<strong>and</strong> <strong>and</strong> also in southern Australia. The sheep from 14 herds in New SouthWales from which the agent (serotypes 2, 3) was isolated had diarrhea <strong>and</strong> someshowed delayed growth <strong>and</strong> died (Philbey et al., 1991). In Great Britain, Y. enterocoliticawas thought <strong>to</strong> have caused abortions in sheep. The etiologic agent was isolatedfrom sheep fetuses <strong>and</strong> most of the serotypes were 6,30 <strong>and</strong> 7, which did nothave the plasmid that determines the markers <strong>to</strong> which virulence is attributed(Corbel et al., 1990). An O:6,30 strain isolated from the liver of an aborted sheepfetus was inoculated intravenously in a group of sheep that had been pregnant forapproximately 90 days; the infection produced a necrotizing placentitis <strong>and</strong> abortions(Corbel et al., 1992).Abortions have been described in association with Y. enterocolitica in cattle in theformer Soviet Union <strong>and</strong> Great Britain <strong>and</strong> in buffalo in India. In the latter country,serotype O:9 was isolated from nine buffaloes that aborted; this serotype shares<strong>common</strong> antigens with Brucella <strong>and</strong> gives serologic cross reactions with that bacterialspecies (Das et al., 1986).Serotypes of Y. enterocolitica were isolated from 5.5% of 451 dogs in Japan(Kaneko et al., 1977) <strong>and</strong> from 1.7% of 115 dogs in Denmark (Pedersen <strong>and</strong>Winblad, 1979). In contrast, the incidence of canine carriers in the US <strong>and</strong> Canadais low. The disease seems <strong>to</strong> occur rarely in dogs, but it should be borne in mind thatmany clinical cases are not diagnosed because isolation is not attempted. In twocases of enteritis described in Canada, the dogs manifested neither fever nor abdominalpains, but they had frequent defecations covered with mucous <strong>and</strong> blood(Papageorges <strong>and</strong> Gosselin, 1983). Y. enterocolitica has also been isolated fromapparently healthy cats. Serotypes O:8 <strong>and</strong> O:9 are among the types isolated fromdogs <strong>and</strong> cats.Infection caused by Y. enterocolitica has been confirmed in several monkeyspecies. In one colony of patas monkeys (Erythrocebus patas) in Missouri (USA),two monkeys died less than one month apart from a generalized infection caused byY. enterocolitica. The remaining 20 monkeys were examined <strong>and</strong> the agent was isolatedfrom rectal swabs taken from five of the clinically normal monkeys (Skavlenet al., 1985).Source of Infection <strong>and</strong> Mode of Transmission (Figure 10): The epidemiologyof enterocolitic yersiniosis is not entirely clear. The agent is widespread in water,food, many animal species, <strong>and</strong> man. Of interest is the fact that the serotypes isolatedfrom water <strong>and</strong> food often do not correspond <strong>to</strong> the types that produce diseasein man. This is also true of the serotypes found in the majority of animal species,


ENTEROCOLITIC YERSINIOSIS 127Figure 10. Enterocolitic yersiniosis (Yersinia enterocolitica). Supposed mode of transmission.SwineFeces, environmental contaminationSwineIngestion of pork productsManFecal-oral routeManwith the exception of pigs <strong>and</strong>, <strong>to</strong> some extent, dogs <strong>and</strong> cats. In countries with thehighest incidence of human disease, pigs are frequently carriers of serotypes pathogenicfor man. In contrast, in those countries where the incidence of human diseaseis low, such as the US or Great Britain, serotypes pathogenic for man are rarely isolatedfrom pigs (Wooley et al., 1980; Brewer <strong>and</strong> Corbel, 1983).Research conducted in Sc<strong>and</strong>inavia, Canada, <strong>and</strong> South Africa strongly suggeststhat the probable reservoir of the agent is swine. In other countries, however, thereservoir is still unknown. Serotype 8, which predominated in the United States, wasisolated from 2 of 95 asymp<strong>to</strong>matic individuals after an outbreak in New York Statedue <strong>to</strong> chocolate milk <strong>and</strong> caused by the same serotype. Serotype 8 was isolatedfrom water <strong>and</strong> foods in the former Czechoslovakia, but no human cases were seen(Aldova et al., 1981). Some nosocomial outbreaks indicate that human-<strong>to</strong>-humantransmission is possible. A study was conducted in a university hospital in theUnited States between 1987 <strong>and</strong> 1990 <strong>to</strong> evaluate nosocomial transmission of theinfection. Of 18 patients from whom Y. enterocolitica was isolated, 8 acquired theinfection outside the hospital, 5 were infected in the hospital 18 <strong>to</strong> 66 days afterbeing admitted for causes other than gastroenteritis, <strong>and</strong> in 5 cases, the origin couldnot be identified (Cannon <strong>and</strong> Linnemann, 1992). Some familial outbreaks havebeen attributed <strong>to</strong> exposure <strong>to</strong> dogs. Nevertheless, dogs <strong>and</strong> cats are not consideredimportant reservoirs.At present, serotype O:3 predominates throughout the world as a humanpathogen. Swine are the primary reservoir <strong>and</strong> source of infection. In Denmark,where there are approximately 2,000 human cases each year, it was demonstratedthat more than 80% of swine herds are infected. Healthy pigs show a high prevalenceof Y. enterocolitica serotype O:3. A study conducted at a slaughterhouse in


128 BACTERIOSESDenmark examined 1,458 pigs; serotype O:3 was isolated from the feces of 360(24.7%) animals. Fecal contamination of the carcass varied with the eviscerationtechnique. In the manual procedure, which is the traditional technique, frequencywas 26.3%, while in the mechanical procedure—suggested along with plugging theanus <strong>and</strong> rectum with a plastic bag—it fell by 1% <strong>to</strong> 2.2%, depending on the regionof the carcass (Andersen, 1988). The clinically important serotypes, O:3, O:5,27,<strong>and</strong> O:8, have been isolated from chopped pork, pig <strong>to</strong>ngue, <strong>and</strong> from chicken.Swine slaughterhouse workers are an occupational group at risk of being infected.Enzyme-linked immunosorbent assay (ELISA) was used <strong>to</strong> examine serum samplesfrom 146 workers in Finl<strong>and</strong>; antibodies were found for serotype O:3 in 19% ofthem <strong>and</strong> in 10% of blood donors used as controls. The <strong>to</strong>nsils of 31 of 120 pigsfrom the same slaughterhouse yielded positive cultures for serotype O:3 (Merilahti-Palo et al., 1991). In a similar study conducted in Norway, 25 (11.1%) of 316slaughterhouse workers <strong>and</strong> 9.9% of 171 veterinarians were positive for IgG antibodies<strong>to</strong> serotype O:3. Counter <strong>to</strong> expectations, of 813 army recruits, prevalencewas higher among those from urban areas (15.2%) than from rural areas (Nesbakkenet al., 1991).Milk <strong>and</strong> water are vehicles of infection, among others. An outbreak in 1976 wasattributed <strong>to</strong> pasteurized chocolate milk. Another outbreak occurred in New York in1981, when 239 people became ill. The largest outbreak of all occurred in severalU.S. states <strong>and</strong> affected 1,000 people who drank recontaminated pasteurized milk.Unlike other outbreaks, the infection was caused by very rare serotypes (O:13a,O:13b). Pasteurization is effective in destroying the agent, <strong>and</strong> thus it is assumedthat contamination occurred afterward. Water contaminated by animal fecal matterhas been assumed <strong>to</strong> be the <strong>common</strong> source of infection in various Nordic countriesin Europe <strong>and</strong> in the US. A small familial outbreak occurred in Canada; it wascaused by serotype O:3, which is responsible for about 75% of all human cases inthat country. The agent was isolated from two family members <strong>and</strong> from water froma shallow well that may have been contaminated by dog feces swept in by heavyrains. The strains from the patients <strong>and</strong> the water had the same characteristics(Thompson <strong>and</strong> Gravel, 1986).It is often difficult <strong>to</strong> identify the source of infection. Foods may contain a smallnumber of pathogenic Y. enterocolitica within a large population of other bacteria,primarily environmental species of Yersinia spp. <strong>and</strong> nonpathogenic serotypes of Y.enterocolitica. Isolation <strong>and</strong> enrichment procedures are not always able <strong>to</strong> detect theetiologic agent (Schiemann, 1989).Blood transfusion is another route for human-<strong>to</strong>-human transmission. Althoughrare, the consequences of such cases are usually serious. From April 1987 <strong>to</strong>February 1991, there were 10 cases in the US of bacteremia caused by transfusionof red blood cells. The final six of these patients showed fever <strong>and</strong> hypotensionwithin 50 minutes of transfusion. One patient suffered explosive diarrhea within 10minutes of transfusion. Four of the six died within a period of 12 hours <strong>to</strong> 37 days.The serotypes isolated were O:5,27 (4 cases), O:3 (one case), <strong>and</strong> O:20 (one case)(CDC, 1991). Blood donors were interviewed <strong>and</strong> some acknowledged having haddiarrhea in the 30 days prior <strong>to</strong> donating blood; one had diarrhea the same day <strong>and</strong>two indicated they had had no gastrointestinal complaints. In Great Britain, four ofa <strong>to</strong>tal of six cases were fatal in 1988. Two cases occurred in Scotl<strong>and</strong> in four monthsalone <strong>and</strong> both people died (Prentice, 1992; Jones et al., 1993). Prentice (1992) esti-


ENTEROCOLITIC YERSINIOSIS 129mates that outside Great Britain there have been 27 cases of septicemia caused byblood transfusion, 17 of them fatal. One case of au<strong>to</strong>logous transfusion has also beendescribed (Richards et al., 1992).The mode of transmission is not well known either, but it is widely accepted thatthe infection is contracted by ingestion of contaminated foods, as in the case of otherenterobacterial <strong>diseases</strong>, as well as by contact with carrier animals <strong>and</strong> by human<strong>to</strong>-humantransmission. It is known that Y. enterocolitica can multiply at refrigerationtemperature. It is believed that this led <strong>to</strong> the 1982 epidemic in the US (see thesection on occurrence in man) produced by recontaminated pasteurized milk. Thisepidemic also reveals that serotypes other than 3, 5, 8, <strong>and</strong> 9 can give rise <strong>to</strong> the disease,although less <strong>common</strong>ly.Role of Animals in the Epidemiology of the Disease: Although not providingdefinitive proof, the accumulated data in countries with a high incidence of thehuman disease indicate that swine are probably an important reservoir of Y. enterocolitica,particularly serotype O:3, which is currently the prevalent type, <strong>and</strong> typeO:9, which is also frequent in swine. The disease caused by a dish prepared with pigintestines (“chitterlings”) in various American cities is good evidence that the infectionis transmitted through food.Diagnosis: In cases of enteritis, appendicitis, erythema nodosum, <strong>and</strong> reactivearthritis, the possibility of Y. enterocolitica infection should be considered. Theagent can be isolated from the patients’ feces. MacConkey agar <strong>and</strong> a selective agarcalled cefsulodina irgasan novobiocin (CIN), created specifically for Yersinia, canbe used for this purpose. Both biotype <strong>and</strong> serotype should be identified. The coldenrichment technique is useful, particularly in the case of carriers that may excretefew Y. enterocolitica cells. Samples are suspended in pep<strong>to</strong>ne culture broth or abuffered phosphate solution for 3 <strong>to</strong> 7 days at 4°C <strong>to</strong> encourage the growth of Y.enterocolitica <strong>and</strong> suppress that of other bacteria. However, routine diagnosis is animpractical procedure, takes a long time (about 1 month), <strong>and</strong> does not exclude nonpathogenicYersinia.Tube serum agglutination <strong>and</strong> the ELISA test can be used with good results asadditional diagnostic techniques. Active infections produce high titers that declineover time. Serum agglutination titers of 1/40 <strong>to</strong> 1/80 are rare in healthy individuals,but <strong>common</strong> in yersiniosis patients <strong>and</strong> can rise <strong>to</strong> very high titers. Positive cultureswithout clear evidence of gastroenteritis are not always accompanied by a highserum agglutination titer.Very high titers are <strong>common</strong> in patients with acute appendicitis (Schiemann, 1989).In countries where serotype 9 is a frequent pathogen for man <strong>and</strong> is also harbored byswine, cross-reaction between Brucella <strong>and</strong> that serotype may cause difficulties.Antibodies in swine against serotype 9 of Y. enterocolitica can be differentiatedfrom those against Brucella by flagellar antigens, which Y. enterocolitica has <strong>and</strong>brucellae do not. Y. enterocolitica also possesses the <strong>common</strong> enterobacterial antigen,which Brucella does not have <strong>and</strong> which therefore may also be used <strong>to</strong> distinguishthem (Mittal et al., 1984). Other animals that have been exposed <strong>to</strong> serotype9 can also show cross-reactions with Brucella.A comparison of three tests for serum diagnosis of type O:3 (immunoelectrophoresis,ELISA, <strong>and</strong> agglutination) produced similar results in terms of sensitivity<strong>and</strong> specificity (Paerregaard et al., 1991).


130 BACTERIOSESA method has been developed for direct identification of Yersinia enterocoliticain blood using polymerase chain reaction (PCR). This procedure can detect theagent in 500 bacteria per 100 microliters of blood (Feng et al., 1992).Control: Currently recommended measures are <strong>to</strong> observe food hygiene rules; <strong>to</strong>ensure that animal products, particularly pork, are well cooked; <strong>and</strong> <strong>to</strong> not drink rawmilk or water of doubtful purity.An important step in prevention is <strong>to</strong> avoid contaminating swine carcasses withfecal matter (see the section on source of infection <strong>and</strong> mode of transmission). Giventhe possibility of interhuman infection in hospitals, generally recommended measuresfor nosocomial infections should be implemented.A practical method for preventing transmission through transfusions is <strong>to</strong> screenwith hema<strong>to</strong>logy stains (Wright, Wright-Giemsa) any blood bank unit that has beenrefrigerated for 25 days or more. Testing has shown that when the contamination isfrom a single colony forming unit (CFU) of Y. enterocolitica, the bacterial count at26 days rises <strong>to</strong> 10 7 –10 8 CFU <strong>and</strong> ≥ 1 ng/mL of endo<strong>to</strong>xin is detected (CDC, 1991).BibliographyAldova, E., J. Sobotková, A. Brezinova, J. Cerna, M. Janeckova, J. Pegrimkova, et al.Yersinia enterocolitica in water <strong>and</strong> foods. Zbl Bakt Mikrobiol Hyg [B] 173:464–470, 1981.Andersen, J.K. Contamination of freshly slaughtered pig carcasses with human pathogenicYersinia enterocolitica. Int J Food Microbiol 7:193–202, 1988.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bissett, M.L., C. Powers, S.L. Abbott, J.M. J<strong>and</strong>a. Epidemiologic investigations of Yersiniaenterocolitica <strong>and</strong> related species: Sources, frequency <strong>and</strong> serogroup distribution. J ClinMicrobiol 28:910–912, 1990.Blumberg, H.M., J.A. Kiehlbauch, I.K. Wachsmuth. Molecular epidemiology of Yersiniaenterocolitica O:3 infections: Use of chromosomal DNA restriction fragment length polymorphismsof rRNA genes. J Clin Microbiol 29:2368–2374, 1991.Brewer, R.A., M.J. Corbel. Characterization of Yersinia enterocolitica strains isolated fromcattle, sheep <strong>and</strong> pigs in the United Kingdom. J Hyg 90:425–433, 1983.Cannon, C.G., C.C. Linnemann. Yersinia enterocolitica infections in hospitalized patients:The problem of hospital-acquired infections. Infect Control Hosp Epidemiol 13:139–143, 1992.Carniel, E., H.H Mollaret. Yersiniosis. Comp Immunol Microbiol Infect Dis 13(2):51–58, 1990.Constantiniu, S., C. Naciu, A. Romaniuc, et al. Serological diagnosis in human Yersiniainfections. Roum Arch Microbiol Immunol 51:225–232, 1992.Corbel, M.J., R.A. Brewer, D. Hunter. Characterisation of Yersinia enterocolitica strainsassociated with ovine abortion. Vet Rec 127:526–527, 1990.Corbel, M.J., B. Ellis, C. Richardson, R. Bradley. Experimental Yersinia enterocolitica placentitisin sheep. Brit Vet J 148:339–349, 1992.Cover, T.L., R.C. Aber. Yersinia enterocolitica. N Engl J Med 321:16–24, 1989.Das, A.M., V.L. Paranjape, S. Winblad. Yersinia enterocolitica associated with thirdtrimester abortion in buffaloes. Trop Anim Health Prod 18:109–112, 1986.de Groote, G., J. V<strong>and</strong>epitte, G. Wauters. Surveillance of human Yersinia enterocoliticainfections in Belgium: 1963–1978. J Infect 4:189–197, 1982.


ENTEROCOLITIC YERSINIOSIS 131Farmer, J.J., III., M.T. Kelly. Enterobacteriaceae. In: Ballows, A., W.J. Hausler, Jr., K.L.Hermann, H.D. Isenberg, H.J. Shadomy. Manual of Clinical Microbiology. 5th ed.Washing<strong>to</strong>n, D.C.: American Society for Microbiology; 1991.Feng, P., S.P. Keasler, W.E. Hill. Direct identification of Yersinia enterocolitica in blood bypolymerase chain reaction amplification. Transfusion 32:850–854, 1992.Franco-Vicario, R., P. Echevarria Villegas, P. Martínez-Olaizola, et al. Yersiniosis en unhospital general del País Vasco (1984–1989). Aspec<strong>to</strong>s clínicos <strong>and</strong> epidemiológicos. MedClin 97:241–244, 1991.Fukushima, H., R. Nakamura, Y. I<strong>to</strong>, K. Sai<strong>to</strong>, M. Tsubokura, K. Otsuki. Ecological studiesof Yersinia enterocolitica. I. Dissemination of Y. enterocolitica in pigs. Vet Microbiol8:469–483, 1983.Hurvell, B. Zoonotic Yersinia enterocolitica infection: Host-range, clinical manifestations,<strong>and</strong> transmission between animals <strong>and</strong> man. In: Bot<strong>to</strong>ne, E.J., ed. Yersinia enterocolitica.Boca Ra<strong>to</strong>n: CRC Press; 1981.Jones, B.L., M.H. Saw, M.F. Hanson, et al. Yersinia enterocolitica septicaemia from transfusionof red cell concentrate s<strong>to</strong>red for 16 days. J Clin Pathol 46:477–478, 1993.Kaneko, K., S. Hamada, E. Ka<strong>to</strong>. Ocurrence of Yersinia enterocolitica in dogs. Jpn J Vet Sci39:407–414, 1977.Lee, L.A., J. Taylor, G.P. Carter, et al. Yersinia enterocolitica O:3: An emerging cause ofpediatric gastroenteritis in the United States. The Yersinia enterocolitica Collaborative StudyGroup. J Infect Dis 163:660–663, 1991.Lindholm, H., R. Visakorpi. Late complications after a Yersinia enterocolitica epidemic: Afollow up study. Ann Rheum Dis 50:694–696, 1991.Marasco, W.J., E.K. Fishman, J.E. Kuhlman, R.H. Hruban. Splenic abscess as a complicationof septic yersinia: CT evaluation. Clin Imaging 17:33–35, 1993.Mata, L., A. Simhon. Enteritis y colitis infecciosas del hombre. Adel Microbiol Enf Infec(Buenos Aires) 1:1–50, 1982.Merilahti-Palo, R., R. Lahesmaa, K. Granfors, et al. Risk of Yersinia infection amongbutchers. Sc<strong>and</strong> J Infect Dis 23:55–61, 1991.Mittal, K.R., I.R. Tizard, D.A. Barnum. Serological cross-reactions between Brucella abortus<strong>and</strong> Yersinia enterocolitica 09. International Symposium on Human <strong>and</strong> AnimalBrucellosis. Taipei, Taiwan, 1984.Morris, J.G., V. Prado, C. Ferreccio, et al. Yersinia enterocolitica isolated from two cohortsof young children in Santiago, Chile: Incidence of <strong>and</strong> lack of correlation between illness <strong>and</strong>proposed virulence fac<strong>to</strong>rs. J Clin Microbiol 29:2784–2788, 1991.Nesbakken, T., G. Kapperud, J. Lassen, E. Skjerve. Yersinia enterocolitica O:3 antibodiesin slaughterhouse employees, veterinarians, <strong>and</strong> military recruits. Occupational exposure <strong>to</strong>pigs as a risk fac<strong>to</strong>r for yersiniosis. Contr Microbiol Immunol 12:32–39, 1991.Paerregaard, A., G.H. Sh<strong>and</strong>, K. Gaarslev, F. Espersen. Comparison of crossed immunoelectrophoresis,enzyme-linked immunosorbent assays, <strong>and</strong> tube agglutination for serodiagnosisof Yersinia enterocolitica serotype O:3 infection. J Clin Microbiol 29:302–309, 1991.Papageorges, M., R. Higgins, Y. Gosselin. Yersinia enterocolitica enteritis in two dogs. JAm Vet Med Assoc 182:618–619, 1983.Pedersen, K.B., S. Winblad. Studies on Yersinia enterocolitica isolated from swine <strong>and</strong>dogs. Acta Path Microbiol Sc<strong>and</strong> [B] 87B:137–140, 1979.Philbey, A.W., J.R. Glas<strong>to</strong>nbury, I.J. Links, L.M. Mathews. Yersinia species isolated fromsheep with enterocolitis. Aust Vet J 68:108–110, 1991.Prentice, M. Transfusing Yersinia enterocolitica. Brit Med J 305:663–664, 1992.Richards, C., J. Kolins, C.D. Trindade. Au<strong>to</strong>logous transfusion-transmitted Yersinia enterocolitica[letter]. JAMA 268:154, 1992.Riley, G., S. Toma. Detection of pathogenic Yersinia enterocolitica by using Congo redmagnesiumoxalate agar medium. J Clin Microbiol 27:213–214, 1989.


132 BACTERIOSESSaebo, A., J. Lassen. Acute <strong>and</strong> chronic liver disease associated with Yersinia enterocoliticainfection: A Norwegian 10-year follow-up study of 458 hospitalized patients. J Intern Med231:531–535, 1992a.Saebo, A., J. Lassen. Acute <strong>and</strong> chronic pancreatic disease associated with Yersinia enterocoliticainfection: A Norwegian 10-year follow-up study of 458 hospitalized patients. J InternMed 231:537–541, 1992b.Schiemann, D.A. Yersinia enterocolitica <strong>and</strong> Yersinia pseudotuberculosis. In: Doyle, M.P.,ed. Foodborne Bacterial Pathogens. New York: Marcel Dekker; 1989.Shayegani, M., W.B. S<strong>to</strong>ne, I. DeForge, et al. Yersinia enterocolitica <strong>and</strong> related speciesisolated from wildlife in New York State. Appl Environ Microbiol 52:420–424, 1986.Skavlen, P.A., H.F. Stills, E.K. Steffan, C.C. Middle<strong>to</strong>n. Naturally ocurring Yersinia enterocoliticasepticemia in patas monkeys (Erythrocebus patas). Lab Anim Sci 35:488–490, 1985.Swaminathan, B., M.C. Harmon, I.J. Mehlman. Yersinia enterocolitica. J Appl Bacteriol52:151–183, 1982.Tacket, C.O., J.P. Narain, R. Sattin, J.P. Lofgren, C. Konigsberg, R.C. Rend<strong>to</strong>rff, et al. Amultistate outbreak of infections caused by Yersinia enterocolitica transmitted by pasteurizedmilk. J Am Med Assoc 251:483–486, 1984.Taylor, D.J. Infection with Yersinia. In: Leman, A.D., B.E. Straw, W.L. Mengeling,S. D’Allaire, D.J. Taylor, eds. Diseases of Swine. 7th ed. Ames: Iowa State UniversityPress; 1992.Thompson, J.S., M.J. Gravel. Family outbreak of gastroenteritis due <strong>to</strong> Yersinia enterocoliticaserotype O:3 from well water. Can J Microbiol 32:700–701, 1986.Trimnell, A.R., A.A. Adesiyun. Characteristics of the first isolate of Yersinia enterocoliticaserogroup O:8 from a dog in Nigeria. Isr J Vet Med 44:244–247, 1988.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Update: Yersinia enterocolitica bacteremia <strong>and</strong> endo<strong>to</strong>xinshock associated with red blood cell transfusions—United States, 1991. MMWR Morb MortalWkly Rep 40(11):176–178, 1991.<strong>WHO</strong> Scientific Working Group. Enteric infections due <strong>to</strong> Campylobacter, Yersinia,Salmonella, <strong>and</strong> Shigella. Bull World Health Organ 58:519–537, 1980.Wooley, R.E., E.B. Shotts, J.W. McConnell. Isolation of Yersinia enterocolitica fromselected animal species. Am J Vet Res 41:1667–1668, 1980.Zamora, J., O. Alonso, E. Chahuán. Isolement et caracterisation de Yersinia enterocoliticachez les rongeurs sauvages du Chili. Zentralbl Veterinarmed [B] 26:392–396, 1979.ENTEROCOLITIS DUE TO CLOSTRIDIUM DIFFICILEICD-10 A04.7Synonyms: Pseudomembranous enterocolitis, antibiotic-associated diarrhea,hemorrhagic necrotizing enterocolitis.Etiology: Clostridium difficile is an anaerobic, gram-positive bacillus 3–16microns long <strong>and</strong> 0.5–1.9 microns in diameter that forms oval, subterminal spores.


ENTEROCOLITIS DUE TO CLOSTRIDIUM DIFFICILE 133Some strains produce chains of two <strong>to</strong> six cells. C. difficile is generally motile inbroth cultures.C. difficile produces two types of <strong>to</strong>xins: entero<strong>to</strong>xin A <strong>and</strong> cy<strong>to</strong><strong>to</strong>xin B. Toxin Ais lethal <strong>to</strong> hamsters when administered orally. Toxin B is cy<strong>to</strong><strong>to</strong>xic for cultured cellsof all types. A picogram of <strong>to</strong>xin B is enough <strong>to</strong> produce the cy<strong>to</strong><strong>to</strong>xic effect (Ca<strong>to</strong>et al., 1986). Not all strains produce <strong>to</strong>xins. Another virulence fac<strong>to</strong>r is a substancethat affects intestinal motility.Various subclassification schemes have been devised for a better underst<strong>and</strong>ing ofthe pathogenicity of C. difficile as well as for epidemiological purposes. One ofthem is based on electrophoretic patterns of proteins on the cellular surface due <strong>to</strong>the different protein profiles produced by SDS-PAGE (polyacrylamide gel electrophoresiswith sodium dodecyl sulphate), staining <strong>and</strong> au<strong>to</strong>radiography ofradiomarked proteins. This method has made it possible <strong>to</strong> distinguish 15 types ofC. difficile (Tabaqchali, 1990). In addition, 15 serogroups were distinguished usingthe plate serotyping system. Six of these serogroups proved <strong>to</strong> be cy<strong>to</strong><strong>to</strong>xigenic. Thecultures were isolated from patients who had pseudomembranous colitis or antibiotic-associateddiarrhea (Toma et al., 1988).Geographic Distribution: Probably worldwide. The agent has been isolatedfrom several sources, such as soil; marine sediment; <strong>and</strong> fecal matter from dogs,cats, cattle, camels, horses <strong>and</strong> other animals; as well as from people withoutdiarrhea (Ca<strong>to</strong> et al., 1986). The number of animals <strong>and</strong> environmental samples(non-nosocomial) studied <strong>to</strong> determine C. difficile carriage was very limited(Levett, 1986).Occurrence in Man: The disease appears sporadically <strong>and</strong> in nosocomial outbreaks.Most cases of pseudomembranous colitis are nosocomial infections (Lyerlyet al., 1988). It is estimated that more than 90% of pseudomembranous colitis casesare due <strong>to</strong> C. difficile <strong>and</strong> that about 20% of diarrhea cases are associated withantibiotics.Occurrence in Animals: Outbreaks of enterocolitis have occurred in horses, rabbits,hamsters, guinea pigs, <strong>and</strong> dogs.In Australia, a study was done of dogs <strong>and</strong> cats treated in two veterinary clinics.C. difficile was successfully cultured in 32 of 81 fecal samples (39.5%). Of the 29animals that received antibiotics, 15 (52%) tested positive in cultures for C. difficile.There was no difference in carriage rate between dogs <strong>and</strong> cats. The environment ofboth clinics was also surveyed for contamination. In one clinic, 15 of 20 sites werecontaminated; in the other, 6 of 14 sites were contaminated. There were both cy<strong>to</strong><strong>to</strong>xigenic<strong>and</strong> noncy<strong>to</strong><strong>to</strong>xigenic isolates. Fifty percent of the animal isolates <strong>and</strong>71.4% of the environmental isolates were not cy<strong>to</strong><strong>to</strong>xigenic. Both dogs <strong>and</strong> cats maybe potential reservoirs (Riley et al., 1991).The Disease in Man: C. difficile produces pseudomembranous colitis or antibiotic-associateddiarrhea in man. The clinical symp<strong>to</strong>ms range from watery diarrhea,with varying degrees of abdominal pain, <strong>to</strong> pseudomembranous hemorrhagic necrotizingcolitis. Infections outside the intestine caused by C. difficile are less important<strong>and</strong> occur less frequently. Abscesses, wound infections, pleurisy, <strong>and</strong> other organiceffects have been described. Arthritis may also occur as a complication of acute colitiscaused by C. difficile (Limonta et al., 1989).


134 BACTERIOSESForty <strong>to</strong> fifty percent of infants have a high load of C. difficile in their intestines,with a high rate of A <strong>and</strong> B <strong>to</strong>xins; despite this, they do not become ill. There is nosatisfac<strong>to</strong>ry explanation for this phenomenon yet (Lyerly et al., 1988). Children whosuffer from other <strong>diseases</strong> or have undergone surgery are at risk of developingpseudomembranous colitis (Adler et al., 1981). In contrast, C. difficile is part of thenormal flora in a very small percentage (approximately 3%) of adults (Limonta etal., 1989).Pseudomembranous colitis was described in the late 1800s, but its importancewas established in the 1970s with the use of antibiotics against anaerobes. Pseudomembranouscolitis emerged as reports began <strong>to</strong> appear on the death of patientstreated with clindamycin, a derivative of lincomycin that had proven effectiveagainst serious anaerobe infections. Diarrhea had already been observed in patientstreated with lincomycin, but with the new antibiotic a severe inflammation of themucosa of the colon with pseudomembranes occurred as well. The mortality rateamong patients treated sometimes reached 10%, but was generally less (Lyerle etal., 1988). It was soon seen that other antibiotics, such as ampicillin <strong>and</strong> cephalosporins,could cause enterocolitis as well (George, 1984). In essence, antibioticsaltered the normal flora of the intestine, disturbing the balance among the differentbacterial species <strong>and</strong> allowing C. difficile <strong>to</strong> multiply.The first step in treatment should be <strong>to</strong> s<strong>to</strong>p treatment with the antibiotic that mayhave caused the disease. The most <strong>common</strong> treatment is with vancomycin, which isnot absorbed in the intestine <strong>and</strong> can reach high concentrations. The patient recoversrapidly. Metronidazole, which is less expensive <strong>and</strong> widely used in Europe, isalso effective. It should be kept in mind that vancomycin <strong>and</strong> metronidazole may, inturn, cause the disease if their concentration in the colon is below an inhibi<strong>to</strong>ry level(Lyerly et al., 1988). Relapses occur in approximately 20% of the patients treated.One study compared the efficacy of vancomycin <strong>and</strong> teicoplanin. Clinical cure wasachieved in 100% of 20 patients treated with vancomycin; 96.2% of 25 patientstreated with teicoplanin were cured. After treatment, five (25%) of those treated withvancomycin <strong>and</strong> two (7.7%) of those treated with teicoplanin were carriers of C. difficile(de Lalla et al., 1992).The Disease in Animals: The difference between the disease in humans <strong>and</strong> animalslies in the different sites affected. While the disease appears primarily as enterocolitisin man, in animals the disease may be cecitis or ileocecitis. Typhlocolitisalso occurs.In the state of Missouri (USA), an outbreak of colitis associated with the possiblyaccidental contamination of feed by lincomycin was described. Seven horses developeddiarrhea. Au<strong>to</strong>psy of a stallion revealed that the cecum was black <strong>and</strong> containedsome 20–30 L of a serosanguineous fluid; the abdominal cavity containedsome 5 L of a clear liquid. Two other outbreaks affecting 15 horses occurred in thesame state (Raisbeck et al., 1981).An outbreak of diarrhea in colts 2 <strong>to</strong> 5 days old occurred in Colorado (USA). C.difficile was isolated from the feces of 27 of 43 neonates with diarrhea (63%) <strong>and</strong>the cy<strong>to</strong><strong>to</strong>xin was detected in the feces of 65% of the animals. C. difficile could notbe isolated from healthy foals <strong>and</strong> adults. This outbreak was not associated withantimicrobial treatment. One foal that died had hemorrhagic necrotizing enteritis; anabundant culture was obtained from the contents of the small intestine (Jones et al.,


ENTEROCOLITIS DUE TO CLOSTRIDIUM DIFFICILE 1351987). Hemorrhagic necrotizing enteritis in neonate foals is usually caused by otherclostridia, such as C. perfringens types B <strong>and</strong> C, <strong>and</strong> C. sordelli. Some cases may bedue <strong>to</strong> C. difficile. C. difficile was isolated from four foals that died at three ranches,<strong>and</strong> the presence of the cy<strong>to</strong><strong>to</strong>xin was also confirmed (Jones et al., 1988). A case oftyphlocolitis was also described in an adult horse (Perrin et al., 1993). Traub-Dargatz <strong>and</strong> Jones (1993) recently reviewed the literature on the disease in horses.Chronic diarrhea due <strong>to</strong> C. difficile was described in dogs; it was successfullytreated with metronidazole (Berry <strong>and</strong> Levett, 1986).A rabbit breeder observed green, watery diarrhea in approximately 25% of his130 animals. Upon au<strong>to</strong>psy, lesions (of varying intensity) were found only in thececum. The <strong>to</strong>tal loss was 40 rabbits. A study confirmed that the feed was contaminatedby a food meant for swine, <strong>to</strong> which lincomycin had been added (permittedonly in feed for pigs <strong>and</strong> fowl). The situation returned <strong>to</strong> normal when the feed waschanged (Thilsted et al., 1981).Hamsters (Mesocricetus auratus) are very susceptible <strong>to</strong> C. difficile <strong>and</strong> are usedas model animals. Proliferative ileitis is seen in young animals; in adult hamsters,the disease is characterized by chronic typhlocolitis with hyperplasia of the mucosa(Rehg <strong>and</strong> Lu, 1982; Chang <strong>and</strong> Rohwer, 1991; Ryden et al., 1991).Outbreaks of typhlitis not induced by antibiotics also occur in guinea pigs. Anoutbreak occurred in a colony of 400 female specific-pathogen free guinea pigs,maintained gno<strong>to</strong>biologically with mice; 123 animals became ill, died, or were sacrificed.The disease was attributed <strong>to</strong> deficient intestinal flora (Boot et al., 1989).Source of Infection <strong>and</strong> Mode of Transmission: Diarrhea due <strong>to</strong> C. difficileoccurs in both man <strong>and</strong> animals absent any association with antibiotics. However,the use of antibiotics <strong>and</strong> the resulting imbalance in the normal intestinal flora is apredominant fac<strong>to</strong>r inducing pseudomembranous enteritis or diarrhea varying fromslight <strong>to</strong> profuse <strong>and</strong> hemorrhagic. The implicated antibiotics are, in particular,clindamycin <strong>and</strong> lincomycin, but other antimicrobials may also be responsible(ampicillin <strong>and</strong> cephalosporins). An intraperi<strong>to</strong>neal injection of ampicillin given <strong>to</strong>mice increased the rate of C. difficile fecal isolates from 19.4% <strong>to</strong> 63.6% (I<strong>to</strong>het al., 1986).The main reservoir of C. difficile seems <strong>to</strong> be infants in the first months of life.The carriage <strong>and</strong> excretion of cy<strong>to</strong><strong>to</strong>xigenic strains in diarrheal dogs may also be anadditional zoonotic source of infection (Berry <strong>and</strong> Levett, 1986; Weber et al., 1989;Riley et al., 1991).Another aspect <strong>to</strong> consider is that C. difficile forms spores that are resistant <strong>to</strong>environmental fac<strong>to</strong>rs. Environmental contamination by C. difficile plays an importantrole in the epidemiology of the disease, in both hamsters <strong>and</strong> man. C. difficilewas isolated from 31.4% of the environmental samples from a hospital ward (Kaatzet al., 1988). Studies conducted with epidemiological markers demonstrate crossinfection between nosocomial patients <strong>and</strong> hospital acquisition of the infection, aswell as a direct relationship between symp<strong>to</strong>ms <strong>and</strong> the type of C. difficile(Tabaqchali, 1990). A recent study on nosocomial transmission is illustrative in thisregard. Rectal swabs taken from 49 chronic-care patients in a geriatric hospital confirmedthe presence of C. difficile in 10 of them (20.4%). A prospective study <strong>to</strong>oksamples from 100 consecutive patients admitted <strong>to</strong> an acute care ward in the samehospital, upon admission <strong>and</strong> every two weeks thereafter. Two patients (2%) were


136 BACTERIOSESpositive upon admission <strong>and</strong> 12 of the initial 98 negatives became colonized by C.difficile, representing a 12.2% nosocomial acquisition rate. The length of hospitalizationwas the most important determinant in colonization (Rudensky et al., 1993).Role of Animals in the Epidemiology of the Disease: Animals play a limitedrole in the transmission of the infection.Diagnosis: Clinical diagnosis of pseudomembranous enterocolitis can beobtained through endoscopy <strong>to</strong> detect the presence of pseudomembranes ormicroabscesses in the colon of diarrheal patients with C. difficile <strong>to</strong>xins in their feces(Lyerly et al., 1988).Labora<strong>to</strong>ry diagnosis consists of culturing the patient’s feces in CCFA medium(cycloserine cefoxitin fruc<strong>to</strong>se egg yolk agar), which is a selective <strong>and</strong> differentialmedium. Patients generally have an elevated number (10 7 or more) of C. difficile intheir feces (Bartlett et al., 1980). The medium can be improved by substitutingsodium taurocholate for egg yolk.Since not all strains are <strong>to</strong>xigenic, detection of the <strong>to</strong>xin in the feces confirms thediagnosis. One of the assays used most often is tissue culture, which is extremelysensitive: it can detect a picogram of <strong>to</strong>xin B (cy<strong>to</strong><strong>to</strong>xin). The mouse lethality testcan also be used. Currently, a commercially available system containing a monolayerof preputial fibroblasts in a 96-well microdilution plate is used (Allen <strong>and</strong>Baron, 1991).Prevention: Avoid abuse of antibiotics. This fac<strong>to</strong>r is particularly acute in thedeveloping countries, where antibiotics can often be obtained without a prescription.Hypochlorite solutions have been recommended for disinfection of surfaces inhospital settings (Kaatz et al., 1988); glutaraldehyde-based disinfectants have beenrecommended for instruments, particularly endoscopes (Rutala et al., 1993).BibliographyAdler, S.P., T. Ch<strong>and</strong>rika, W.F. Berman. Clostridium difficile associated with pseudomembranouscolitis. Occurrence in a 12 week-old infant without prior antibiotic therapy. Am J DisChild 135:820–822, 1981.Allen, S.D., E.J. Baron. Clostridium. In: Ballows, A., W.J. Hausler, K.L. Hermann, H.D.Isenberg, H.J. Shadomy, eds. Manual of Clinical Microbiology. 5th ed. Washing<strong>to</strong>n, D.C.:American Society for Microbiology; 1991.Bartlett, J.G., N.S. Taylor, T. Chang, J. Dzink. Clinical <strong>and</strong> labora<strong>to</strong>ry observations inClostridium difficile colitis. Am J Clin Nutr 33:2521–2526, 1980.Berry, A.P., P.N. Levett. Chronic diarrhoea in dogs associated with Clostridium difficile. VetRec 118:102–103, 1986.Boot, R., A.F. Angulo, H.C. Walvoort. Clostridium difficile-associated typhlitis in specificpathogen free guineapigs in the absence of antimicrobial treatment. Lab Anim 23:203–207, 1989.Ca<strong>to</strong>, E.P., W.L. George, S.M. Finegold. Genus Clostridium Prazmowski 1880. In: Sneath,P.H.A., N.S. Mair, M.E. Sharpe, J.G. Holt. Vol 2: Bergey’s Manual of Systemic Bacteriology.Baltimore: Williams & Wilkins; 1986.Chang, J., R.G. Rohwer. Clostridium difficile infection in adult hamsters. Lab Anim Sci41:548–552, 1991.


ENTEROCOLITIS DUE TO CLOSTRIDIUM DIFFICILE 137de Lalla, F., R. Nicolin, E. Rinaldi, et al. Prospective study of oral teicoplanin versus oralvancomycin for therapy of pseudomembranous colitis <strong>and</strong> Clostridium difficile-associateddiarrhea. Antimicrob Agents Chemother 36:2192–2196, 1992.George, W.L. Antimicrobial agent-associated colitis <strong>and</strong> diarrhea: His<strong>to</strong>rical background<strong>and</strong> clinical aspects. Rev Infect Dis 6(Suppl 1):S208–S213, 1984.I<strong>to</strong>h, K., W.K. Lee, H. Kawamura, et al. Isolation of Clostridium difficile from variouscolonies of labora<strong>to</strong>ry mice. Lab Anim 20:266–270, 1986.Jones, R.L., W.S. Adney, A.F. Alex<strong>and</strong>er, et al. Hemorrhagic necrotizing enterocolitis associatedwith Clostridium difficile infection in four foals. J Am Vet Med Assoc 193:76–79, 1988.Jones, R.L., W.S. Adney, R.K. Shidaler. Isolation of Clostridium difficile <strong>and</strong> detection ofcy<strong>to</strong><strong>to</strong>xin in the feces of diarrheic foals in the absence of antimicrobial treatment. J ClinMicrobiol 25:1225–1227, 1987.Kaatz, G.W., S.D. Gitlin, D.R. Schaberg, et al. Acquisition of Clostridium difficile from thehospital environment. Am J Epidemiol 127:1289–1294, 1988.Limonta, M., A. Arosio, D. Salvioni, A. De Carli. Due casi di artralgia associati adinfezione da Clostridium difficile. Boll Ist Sieroter Milan 68:142–144, 1989.Levett, P.N. Clostridium difficile in habitats other than the human gastrointestinal tract. JInfect 12:253–263, 1986.Lyerly, D.M., H.C. Krivan, T.D. Wilkins. Clostridium difficile: Its disease <strong>and</strong> <strong>to</strong>xins. ClinMicrobiol Rev 1:1–18, 1988.Perrin, J., I. Cosmeta<strong>to</strong>s, A. Galluser, et al. Clostridium difficile associated with typhlocolitisin an adult horse. J Vet Diagn Invest 5:99–101, 1993.Raisbeck, M.F., G.R. Holt, G.D. Osweiler. Lincomycin-associated colitis in horses. J AmVet Med Assoc 179:362–363, 1981.Rehg, J.E., Y.S. Lu. Clostridium difficile typhlitis in hamsters not associated with antibiotictherapy. J Am Med Vet Assoc 181:1422–1423, 1982.Riley, T.V., J.E. Adams, G.L. O’Neill, R.A. Bowman. Gastrointestinal carriage ofClostridium difficile in cats <strong>and</strong> dogs attending veterinary clinics. Epidemiol Infect107:659–665, 1991.Rudensky, B., S. Rosner, M. Sonnenblick, et al. The prevalence <strong>and</strong> nosocomial acquisitionof Clostridium difficile in elderly hospitalized patients. Postgrad Med J 69:45–47, 1993.Rutala, W.A., M.F. Gergen, D.J. Weber. Inactivation of Clostridium difficile spores by disinfectants.Infect Control Hosp Epidemiol 14:36–39, 1993.Ryden, E.B., N.S. Lipman, N.S. Taylor, et al. Clostridium difficile typhlitis associated withcecal mucosal hyperplasia in Syrian hamsters. Lab Anim Sci 41:553–558, 1991.Tabaqchali, S. Epidemiologic markers of Clostridium difficile. Rev Infect Dis 12(Suppl2):S192–S199, 1990.Thilsted, J.P., W.M. New<strong>to</strong>n, R.A. Cr<strong>and</strong>ell, R.F. Bevill. Fatal diarrhea in rabbits resultingfrom the feeding of antibiotic-contaminated feed. J Am Vet Med Assoc 179:360–362, 1981.Toma, S., G. Lesiak, M. Magus, et al. Serotyping of Clostridium difficile. J Clin Microbiol26:426–428, 1988.Traub-Dargatz, J.L., R.L. Jones. Clostridia-associated enterocolitis in adult horses <strong>and</strong>foals. Vet Clin North Am Equine Pract 9:411–421, 1993.Weber, A., P. Kroth, G. Heil. Untersuchungen zum Vorkommen von Clostridium difficile inKotproben von Hunden und Katzen. Zentralbl Veterinarmed 36:568–576, 1989.


138 BACTERIOSESFOOD POISONING CAUSED BYVIBRIO PARAHAEMOLYTICUSICD-10 A05.3 foodborne Vibrio parahaemolyticus in<strong>to</strong>xicationEtiology: Vibrio parahaemolyticus, belonging <strong>to</strong> the family Vibrionaceae, is agram-negative, motile, curved or straight bacillus that does not produce spores. It isa halophile that develops best in media with 2% <strong>to</strong> 3% sodium chloride but can multiplyin an 8% concentration of this salt. In most cases, isolated strains are ureasenegative, but urease-positive strains are also found; this difference may serve as anepidemiological marker.Based on O (somatic) <strong>and</strong> K (capsular) antigens, 20 O groups <strong>and</strong> 65 K serotypesare serologically distinguished. Most clinical strains can be typed, but environmentalstrains cannot.Many clinical strains of V. parahaemolyticus cultured in Wagatsuma agar (whichcontains human red blood cells) are beta-hemolytic, while environmental isolatesfrom water are not. This has been called the Kanagawa phenomenon or test. Giventhe difference in hemolytic capacity between clinical <strong>and</strong> environmental strains, itwas assumed that hemolysin is a virulence fac<strong>to</strong>r. This <strong>to</strong>xin is called thermostabledirect hemolysin (TDH). However, it has been demonstrated that TDH-negativestrains can cause disease <strong>and</strong> produce an immunologically related <strong>to</strong>xin, thermostablerelated hemolysin (TRH), with very similar properties. The twohemolysins are coded by two different genes. In some strains, it was possible <strong>to</strong> findboth hemolysins. Of 112 V. parahaemolyticus strains studied, 52.3% had the TDHgene alone, 24.3% had the TRH gene, <strong>and</strong> 11.2% had both genes (TDH <strong>and</strong> TRH).It can thus be stated that TDH <strong>and</strong> TRH are important fac<strong>to</strong>rs in virulence (Shirai etal., 1990). In addition, strains from diarrhea patients producing TRH were comparedwith environmental strains of V. parahaemolyticus (isolated from seawater orseafood) producing TRH. The results show that they were indistinguishable (Yoh etal., 1992).Pili are another important fac<strong>to</strong>r in intestinal colonization <strong>and</strong> thus in virulence.Various researchers have shown that pili attach <strong>to</strong> rabbit intestinal epithelium <strong>and</strong>that adhesive capacity is blocked by treating the vibrios with anti-pilus antibodies(Fab fraction). This does not produce an antihemolysin serum (Nakasone <strong>and</strong>Iwanaga, 1990 <strong>and</strong> 1992; Chakrabarti et al., 1991).Geographic Distribution: V. parahaemolyticus has been isolated from sea <strong>and</strong>estuary waters on all continents. The agent’s distribution shows marked seasonalvariations in natural reservoirs. During cold months, it is found in marine sediment;during warm months, it is found in coastal waters, fish, <strong>and</strong> shellfish (Benenson,1990). There have been a few reports of the isolation of V. parahaemolyticus fromcontinental waters <strong>and</strong> fish in rivers or lakes. It is assumed that these waters had ahigh concentration of sodium chloride, which would allow the agent <strong>to</strong> survive(Twedt, 1989). The fac<strong>to</strong>rs that determine the abundance of the bacteria includewater temperature, salinity, <strong>and</strong> plank<strong>to</strong>n, among other fac<strong>to</strong>rs.The countries most affected by the disease are Japan, Taiwan, <strong>and</strong> other Asiancoastal regions, though cases of disease have been described in many countries <strong>and</strong>on many continents.


FOOD POISONING CAUSED BY VIBRIO PARAHAEMOLYTICUS 139Occurrence in Man: Food poisoning caused by this agent occurs sporadically orin outbreaks. Much of the knowledge about this disease is due <strong>to</strong> researchers inJapan, where the disease was first described in 1953. Subsequent studies showedthat during the summer months, 50% <strong>to</strong> 70% of food poisoning cases <strong>and</strong> outbreakswere caused by V. parahaemolyticus (Snydman <strong>and</strong> Gorbach, 1991).It is difficult <strong>to</strong> estimate the number of sporadic cases that occur. Many of thosewho fall ill do not see a doc<strong>to</strong>r, <strong>and</strong> if they do, diagnosis is limited <strong>to</strong> a clinical examinationwithout labora<strong>to</strong>ry confirmation. Outbreaks can affect few or many people.During an outbreak that occurred in 1978, two-thirds of the 1,700 people whoattended a dinner in Port Allen, Louisiana (USA) fell ill. The source of the outbreakwas probably undercooked shrimp (CDC, 1978). The attack rate of people exposedduring outbreaks in the US varied from 24% <strong>to</strong> 86% <strong>and</strong> the number of thoseaffected ranged from 6 <strong>to</strong> 600. In the four years between 1983 <strong>and</strong> 1986, there wasan outbreak that affected two people (Snydman <strong>and</strong> Gorbach, 1991).Another outbreak that affected several hundred people occurred in the Bahamasin 1991. At the most critical point in the outbreak, 348 cases were treated in a hospitalon the isl<strong>and</strong>. The outbreak was attributed <strong>to</strong> a gastropod (Strombus gigas),<strong>common</strong>ly called “conch,” that the population usually eats raw or partially cooked.Kanagawa-positive V. parahaemolyticus was isolated from 5 of 14 patients’ s<strong>to</strong>olsamples; two positive cultures were also isolated from eight conch samples.Although the number of cultures was limited, it is thought that V. parahaemolyticuswas the causative agent of the diarrheal disease, which during the entire course ofthe outbreak affected more than 800 people, most of them adults.In British Columbia (Canada), V. parahaemolyticus cultures were isolated from13 patients as well as from 221 environmental samples; 23% <strong>and</strong> 1.4%, respectively,were Kanagawa positive. The cases of infection contracted locally were urease positive<strong>and</strong> Kanagawa negative; the patients who were infected during a trip abroadwere urease negative <strong>and</strong> Kanagawa positive. Eight percent of the environmentalsamples were also urease positive <strong>and</strong> Kanagawa negative. These results suggestthat the hemolysin identified by the Kanagawa test is not the only hemolysininvolved in the pathogenesis of the infection (Kelly <strong>and</strong> Stroh, 1989. Also see thesection on etiology).In Recife, in northeastern Brazil, in 8 (38%) of 21 fecal samples from adultpatients with gastroenteritis, cultures were also isolated that were urease positive<strong>and</strong> Kanagawa negative (Magalhães et al., 1991b). Also in Recife, V. parahaemolyticuswas isolated from 14 (1.3%) of 1,100 diarrheal fecal samples. If onlyadult samples are taken in<strong>to</strong> account, the isolation rate would be 7.1%. It was alsopossible <strong>to</strong> show that the cultures belonged <strong>to</strong> seven different K antigen serovars(Magalhães et al., 1991a).Occurrence in Animals: V. parahaemolyticus is frequently isolated from fish,mollusks, <strong>and</strong> crustaceans in coastal waters, throughout the year in tropical climates<strong>and</strong> during the summer months in cold or temperate climates.The Disease in Man: The incubation period is from 12 <strong>to</strong> 24 hours, but may varyfrom 6 <strong>to</strong> more than 90 hours. The most prominent symp<strong>to</strong>m is watery diarrhea,which becomes bloody in some cases, as has been seen in Bangladesh, the US, <strong>and</strong>India. The other <strong>common</strong> symp<strong>to</strong>ms are abdominal pains, nausea, vomiting, cephalalgia,<strong>and</strong> sometimes fever <strong>and</strong> chills (Twedt, 1989).


140 BACTERIOSESThe disease is usually mild <strong>and</strong> lasts from one <strong>to</strong> seven days, but there have beenfatal cases (Klontz, 1990). Some extraintestinal cases have occurred, such as infectionof wounds, ears, <strong>and</strong> eyes, <strong>and</strong> there have also been isolates from blood. In someof these latter cases, there is doubt as <strong>to</strong> whether they were caused by V. parahaemolyticusor other halogenous Vibrios. Sautter et al. (1988) described the case ofa foot wound infected by Kanagawa-negative V. parahaemolyticus. A hospitalemployee suffered a superficial abrasion <strong>and</strong> a small bruise on the ankle <strong>and</strong> traveledthe following day <strong>to</strong> the eastern coast of the US. The abrasion began <strong>to</strong> ulcerate,edema <strong>and</strong> erythema formed around the ulcer, <strong>and</strong> the area became painful. By thesixth day, the erythema had grown <strong>to</strong> 18 cm <strong>and</strong> a 4 cm ulcer appeared. The patientwas treated with dicloxacillin for 14 days. After two days of treatment, the ulcerbegan <strong>to</strong> leak a serosanguineous fluid, from which V. parahaemolyticus was isolated.Treatment was completed, the patient recovered, <strong>and</strong> the cultures were negative.Generally no treatment other than rehydration is required for food poisoningcaused by V. parahaemolyticus. The use of antibiotics should be reserved for prolongedor severe cases.The Disease in Animals: V. parahaemolyticus causes only an inapparent contaminationor infection in fish, mollusks, <strong>and</strong> crustaceans.Source <strong>and</strong> Mode of Transmission: The major reservoir is seawater. Fish, mollusks,<strong>and</strong> crustaceans acquire the infection from seawater. When humans eat themraw or insufficiently cooked, they act as a source of infection. Humans need a loadof 10 5 –10 7 of V. parahaemolyticus <strong>to</strong> become infected (Twedt, 1989).Recently caught fish have a V. parahaemolyticus load of only 1,000 per gram orless <strong>and</strong> recently harvested mollusks have a load of some 1,100 per gram; i.e., a loadlower than that needed <strong>to</strong> infect humans (Twedt, 1989). It is thus assumed that thehigher load is caused by h<strong>and</strong>ling of these seafoods, permitting multiplication of V.parahaemolyticus in the food. V. parahaemolyticus reproduces in a very short time(approximately 12 minutes) <strong>and</strong> exposure of the food <strong>to</strong> room temperature for a fewhours is enough <strong>to</strong> allow the bacterial load <strong>to</strong> produce poisoning in man.A very important fac<strong>to</strong>r in the epidemiology of the disease in many countries isthe cus<strong>to</strong>m of eating raw seafood. Japan is one of the countries with the most outbreaksof food poisoning caused by V. parahaemolyticus because raw fish, shellfish,<strong>and</strong> crustaceans are consumed there. In the US, the most <strong>common</strong> source of poisoningis the consumption of raw oysters <strong>and</strong> even some uncooked or undercookedcrustaceans.Carrier status lasts for a few days <strong>and</strong> there are no known cases of secondaryinfection.Role of Animals: The role is indirect <strong>and</strong> transmission is through food. The onlyvertebrates involved in the chain of transmission <strong>to</strong> man are fish, along with mollusks<strong>and</strong> crustaceans.Diagnosis: A diarrheal disease occurring during the warm months <strong>and</strong> in associationwith the ingestion of seafood should lead one <strong>to</strong> suspect the possibility of foodpoisoning caused by V. parahaemolyticus. Certain diagnosis is obtained throughisolation <strong>and</strong> characterization of the etiologic agent.The medium most often used for culturing feces is thiosulfate citrate bile saltssucrose (TCBS) agar. The colonies in this medium take on a green or bluish color,


FOOD POISONING CAUSED BY VIBRIO PARAHAEMOLYTICUS 141with a darker green center. As a pre-enrichment medium, water with 1% pep<strong>to</strong>ne <strong>and</strong>3% salt can be used. Wagatsuma medium is used <strong>to</strong> determine whether the cultureis Kanagawa-positive or negative.Prevention: The main recommendation is <strong>to</strong> cook shellfish, crustaceans, <strong>and</strong> fishat a sufficiently high temperature (15 minutes at 70°C) <strong>to</strong> destroy V. parahaemolyticus,with particular attention <strong>to</strong> the volume of the seafood in order <strong>to</strong>achieve the appropriate temperature.However, the well-established cus<strong>to</strong>m in some countries of eating raw seafoodmakes it very difficult <strong>to</strong> enforce the recommendation <strong>to</strong> inactivate V. parahaemolyticusin fish, crustaceans, <strong>and</strong> mollusks by sufficiently cooking these foods.An experiment conducted <strong>to</strong> study the increase of hemolysin in comparison with thebacterial count reached the conclusion that the <strong>to</strong>xin appears when V. parahaemolyticusreaches the level of 10 6 per gram <strong>and</strong> continues <strong>to</strong> increase with multiplication ofthe microbe. At 35°C it reached 32 units of hemolysin per gram after 24 hours; at 25°Cit reached this level after 48 hours. Once formed, hemolysin is quite stable. Hemolysinshowed its maximum heat resistance at a pH of between 5.5 <strong>and</strong> 6.5. The Kanagawahemolysin in shrimp homogenate proved <strong>to</strong> be stable for 17 days when kept at 4°C; attemperatures between 115°C <strong>and</strong> 180°C, it <strong>to</strong>ok between 48.1 <strong>and</strong> 10.4 minutes forthermal inactivation as demonstrated in rats (Bradshaw et al., 1984).The results of this <strong>and</strong> other experiments indicate that from the outset it is necessary<strong>to</strong> prevent the load of V. parahaemolyticus in seafood as much as possible. Acontra-indicated practice is washing fish or other seafood in contaminated estuarinewater. Cold s<strong>to</strong>rage is recommended as soon as possible after cooking. Table surfaceswhere these products are processed should be waterproof <strong>and</strong> must be completelycleaned with fresh water (without salt) as there may be cross contamination,particularly from salted foods.BibliographyBenenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bradshaw, J.G., D.B. Shah, A.J. Wehby, et al. Thermal inactivation of the Kanagawahemolysin of Vibrio parahaemolyticus in buffer <strong>and</strong> shrimp. J Food Sci 49:183–187, 1984.Chakrabarti, M.K., A.K. Sinha, T. Biswas. Adherence of Vibrio parahaemolyticus <strong>to</strong> rabbitintestinal epithelial cells in vitro. FEMS Microbiol Lett 68:113–117, 1991.Doyle, M.P. Pathogenic Escherichia coli, Yersinia enterocolitica, <strong>and</strong> Vibrio parahaemolyticus.Lancet 336(8723):1111–1115, 1990.Kelly, M.T., E.M. Stroh. Urease-positive, Kanagawa-negative Vibrio parahaemolyticusfrom patients <strong>and</strong> the environment in the Pacific Northwest. J Clin Microbiol 27:2820–2822, 1989.Klontz, K.C. Fatalities associated with Vibrio parahaemolyticus <strong>and</strong> Vibrio cholerae non-O1 infections in Florida (1981 <strong>to</strong> 1988). South Med J 83:500–502, 1990.Magalhães, V., R.A. Lima, S. Tateno, M. Malgahães. Gastroenterites humanas associadas aVibrio parahaemolyticus no Recife, Brasil. Rev Inst Med Trop Sao Paulo 33:64–68, 1991a.Magalhães, M., V. Malgahães, M.G. Antas, S. Tateno. Isolation of urease-positive Vibrioparahaemolyticus from diarrheal patients in northeast Brasil. Rev Inst Med Trop Sao Paulo33:263–265, 1991b.


142 BACTERIOSESNakasone, N., M. Iwanaga. Pili of a Vibrio parahaemolyticus strain as a possible colonizationfac<strong>to</strong>r. Infect Immun 58:61–69, 1990.Nakasone, N., M. Iwanaga. The role of pili in colonization of the rabbit intestine by Vibrioparahaemolyticus Na2. Microbiol Immunol 36:123–130, 1992.Sautter, R.L., J.S. Taylor, J.D. Oliver, C. O’Donnell. Vibrio parahaemolyticus (Kanagawanegative)wound infection in a hospital dietary employee. Diagn Microbiol Infect Dis 9:41–45, 1988.Shirai, H., H. I<strong>to</strong>, T. Hirayama, et al. Molecular epidemiologic evidence for association ofthermostable direct hemolysin (TDH) <strong>and</strong> TDH-related hemolysin of Vibrio parahaemolyticuswith gastroenteritis. Infect Immun 58:3568–3573, 1990.Snydman, D.R., S.L. Gorbach. Bacterial food poisoning. In: Evans, A.S., P.S. Brachman,eds. Bacterial Infections of Humans. 2nd ed. New York: Plenum Medical Book Co.; 1991.Spriggs, D.R., R.B. Sack. From the National Institute of Allergy <strong>and</strong> Infectious Diseases.Summary of the 25th United States-Japan Joint Conference on Cholera <strong>and</strong> Related DiarrhealDiseases. J Infect Dis 162:584–590, 1990.Twedt, R.M. Vibrio parahaemolyticus. In: Doyle, M., ed. Foodborne Bacterial Pathogens.New York: Marcel Dekker; 1989.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Vibrio parahaemolyticus foodborne outbreak—Louisiana.MMWR Morb Mortal Wkly Rep 27:345–346, 1978.Yoh, M., T. Miwatani, T. Honda. Comparison of Vibrio parahaemolyticus hemolysin(Vp-TRH) produced by environmental <strong>and</strong> clinical isolates. FEMS Microbiol Lett71:157–161, 1992.


GLANDERSICD-10 A24.0Synonyms: Farcy, cutaneous gl<strong>and</strong>ers, equine nasal phthisis, maliasmus.Etiology: Pseudomonas (Malleomyces, Actinobacillus) mallei, a nonmotile,gram-negative bacillus that is not very resistant <strong>to</strong> environmental conditions; this isthe only nonmotile species in the genus Pseudomonas.Geographic Distribution: At one time, the disease was distributed worldwide. Itwas eradicated in Europe <strong>and</strong> the Americas, but foci reappeared in 1965 in Greece,Romania, <strong>and</strong> Brazil (FAO/<strong>WHO</strong>/OIE, 1972). The present distribution is not wellknown, but there are indications that it persists in some African <strong>and</strong> Asian countries;Mongolia is or was the area of greatest incidence. According <strong>to</strong> official countryreports <strong>to</strong> the Food <strong>and</strong> Agriculture Organization of the United Nations (FAO), theInternational Office of Epizootics (OIE), <strong>and</strong> the World Health Organization(<strong>WHO</strong>), no government currently reports cases of gl<strong>and</strong>ers. Isolated suspected caseswere noted in Mongolia <strong>and</strong> diagnostic tests were being conducted. No cases havebeen reported since 1991 in India <strong>and</strong> since 1987 in Iraq (FAO/<strong>WHO</strong>/OIE, 1992).


GLANDERS 143Occurrence in Man: At present, the disease in man is exceptional, if it occurs atall. Attenuated strains of P. mallei are found in Asia, where the infection is assumed<strong>to</strong> persist.Occurrence in Animals: According <strong>to</strong> various sources, incidence in solipeds isnow low or nonexistent in Myanmar (Burma), China, India, Indonesia, Vietnam, <strong>and</strong>Thail<strong>and</strong>, <strong>and</strong> the disease is seen only occasionally. Cases used <strong>to</strong> occur sporadicallyin Pakistan <strong>and</strong> rarely in Iran. In June 1982, 826 foci with 1,808 cases were reportedin solipeds in Turkey <strong>and</strong> in 1984, 274 foci were reported (OIE, 1982, 1984). The incidencein Mongolia is believed <strong>to</strong> have been high. The present situation in Ethiopia <strong>and</strong>the Central African Republic is not known, but cases have occurred in these countriesin recent years. The most recent information available is from the “GeographicDistribution” section of the Animal Health Yearbook (FAO/<strong>WHO</strong>/OIE, 1993). Basedon the reports obtained, it would seem that the disease is becoming extinct.In endemic areas, the incidence of infection was higher during the rainy season.The Disease in Man: The incubation period is usually from 1 <strong>to</strong> 14 days. Casesof latent infection that became clinically evident after many years have beendescribed. The disease course may be either acute or chronic. In addition, subclinicalinfections have been discovered during au<strong>to</strong>psy.In man as well as in animals, P. mallei tends <strong>to</strong> localize in the lungs, nasal mucosa,larynx, <strong>and</strong> trachea. The infection is manifested clinically as pneumonia, bronchopneumonia,or lobar pneumonia, with or without bacteremia. Pulmonary abscesses,pleural effusion, <strong>and</strong> empyema may occur. In the acute forms, there is mucopurulentdischarge from the nose, <strong>and</strong> in the chronic forms, granuloma<strong>to</strong>us nodular lesionsare found in the lungs.Ulcers appear in the mucosa of the nostrils <strong>and</strong> may also be found in the pharynx.Cellulitis with vesiculation, ulceration, lymphangitis, <strong>and</strong> lymphadenopathy is seenon the skin at the etiologic agent’s point of entry. Mortality in clinical cases is high.The Disease in Animals: Gl<strong>and</strong>ers is primarily a disease of solipeds. The diseasecourse is predominantly chronic in horses <strong>and</strong> is almost always acute in asses <strong>and</strong>mules. The acute form results in high fever, depression, dyspnea, diarrhea, <strong>and</strong> rapidweight loss. The animal dies in a few weeks. The chronic form may last years; someanimals recover, others die.Chronic gl<strong>and</strong>ers is characterized by three clinical forms, occurring alone orsimultaneously: pulmonary gl<strong>and</strong>ers, upper respira<strong>to</strong>ry tract disease, <strong>and</strong> cutaneousgl<strong>and</strong>ers.Pulmonary gl<strong>and</strong>ers can remain inapparent for lengthy periods. When clinicalsymp<strong>to</strong>ms do occur, they consist of intermittent fever, cough, depression, <strong>and</strong> weightloss. In more advanced stages, there is dyspnea with rales. Pulmonary lesions usuallyconsist of nodules or pneumonic foci. The nodules are grayish white with redborders; in time, the center becomes caseous or soft, or undergoes calcification <strong>and</strong>becomes surrounded by grayish granulated or whitish fibrous tissue.The upper respira<strong>to</strong>ry disease is characterized by ulcerations of the mucosa(necrosis of the nodules is the initial lesion) of one or both nostrils <strong>and</strong>, frequently,of the larynx <strong>and</strong> trachea. The ulcers have a grayish center with thick, jagged borders.There is a mucous or mucopurulent discharge from one or both nostrils thatforms dark scabs around them.


144 BACTERIOSESFigure 11. Gl<strong>and</strong>ers. Mode of transmission.InfectedsolipedsContaminated fodder, water<strong>and</strong> objects; direct contact; aerosolsSusceptiblesolipedsIngestion of meatDirect contact (skin);aerosols (nasal <strong>and</strong> conjunctival route)Felidsin zoosManThe cutaneous form (farcy) begins with superficial or deep nodules; these laterbecome ulcers that have a gray center <strong>and</strong> excrete a thick, oily liquid that encruststhe hair. The lymph vessels form visible cords, <strong>and</strong> the lymph nodes are swollen.Most authors consider upper respira<strong>to</strong>ry gl<strong>and</strong>ers <strong>and</strong> cutaneous gl<strong>and</strong>ers <strong>to</strong> besecondary forms of pulmonary gl<strong>and</strong>ers.In zoos <strong>and</strong> circuses, carnivores have contracted gl<strong>and</strong>ers as a consequence of eatingmeat from infected solipeds. The dog is another accidental host.Source of Infection <strong>and</strong> Mode of Transmission (Figure 11): Man contracts theinfection through contact with sick solipeds, especially those kept in crowded conditions,such as army stables. The portals of entry are the skin <strong>and</strong> the nasal <strong>and</strong> ocularmucosa. Nasal discharges, skin ulcer secretions, <strong>and</strong> contaminated objects constitutethe source of infection.Solipeds acquire the infection from conspecifics, mainly via the digestive route,but probably also through inhalation <strong>and</strong> wound infection.Role of Animals in the Epidemiology of the Disease: The reservoir of P. malleiis solipeds. The great epizootics of gl<strong>and</strong>ers have occurred in metropolitan stables,especially during wartime. Horses with chronic or latent infection are responsiblefor maintaining the disease in an establishment or region, <strong>and</strong> their movementfrom one place <strong>to</strong> another contributes <strong>to</strong> its spread. Man <strong>and</strong> carnivores are accidentalhosts.Diagnosis: Diagnosis of gl<strong>and</strong>ers is based on: (a) bacteriologic examinations bymeans of culture or inoculation in<strong>to</strong> hamsters of nasal or skin secretions or tissuefrom internal organs, especially the lungs; (b) allergenic tests with mallein (the intrapalpebraltest is preferred); <strong>and</strong> (c) serologic tests, especially complement fixation.Although this last test is considered specific, false positives have occurred.


GLANDERS 145Control: Prevention in humans consists primarily of eradication of the infectionin solipeds. Greatly improved diagnostic methods have made successful eradicationcampaigns possible, as have the disapperance of stables from cities <strong>and</strong> the almostcomplete substitution of au<strong>to</strong>mobiles for horses. Eradication procedures consist ofidentification of infected animals with allergenic or serologic tests, <strong>and</strong> sacrifice ofreac<strong>to</strong>rs. Installations <strong>and</strong> equipment must then be disinfected.BibliographyBlood, D.C., J.A. Henderson. Veterinary Medicine. 4th ed. Baltimore: Williams &Wilkins; 1974.Bruner, D.W., J.H. Gillespie. Hagan’s Infectious Diseases of Domestic Animals. 6th ed.Ithaca: Coms<strong>to</strong>ck; 1973.Cluff, L.E. Diseases caused by Malleomyces. In: Beeson, P., B.W. McDermott, J.B.Wyngaarden, eds. Cecil Textbook of Medicine. 15th ed. Philadelphia: Saunders; 1979.Food <strong>and</strong> Agriculture Organization of the United Nations (FAO)/World HealthOrganization (<strong>WHO</strong>)/International Office of Epizootics (OIE). Animal Health Yearbook,1971. Rome: FAO; 1972.Food <strong>and</strong> Agriculture Organization of the United Nations (FAO)/World HealthOrganization (<strong>WHO</strong>)/International Office of Epizootics (OIE). Animal Health Yearbook,1984. Rome: FAO; 1985.Food <strong>and</strong> Agriculture Organization of the United Nations (FAO)/World HealthOrganization (<strong>WHO</strong>)/International Office of Epizootics (OIE). Animal Health Yearbook,1992. Rome: FAO; 1993. (FAO Production <strong>and</strong> Animal Health Series 32).Hagebock, J.M., L.K. Schlater, W.M. Frerichs, D.P. Olson. Serologic responses <strong>to</strong> themallein test for gl<strong>and</strong>ers in solipeds. J Vet Diagn Invest 5:97–99, 1993.Hipóli<strong>to</strong>, O., M.L.G. Freitas, J.B. Figuereido. Doenças Infe<strong>to</strong>-Contagiosas dos AnimaisDomésticos. 4th ed. São Paulo: Melhoramen<strong>to</strong>s; 1965.International Office of Epizootics (OIE). Report on the Disease Status Worldwide in 1984.53rd General Session. Paris: IOE; 1985. (Document 53SG/2).International Office of Epizootics (OIE). Enfermedades animales señaladas a la OIE,estadísticas 1982. Paris: OIE; 1982.Langeneger, J., J. Dobereiner, A.C. Lima. Foco de mormo (Malleus) na região de Campos,Estado do Rio de Janeiro. Arq Inst Biol Anim 3:91–108, 1960.Oudar, J., L. Dhennu, L. Joubert, A. Richard, J.C. Coutard, J.C. Proy, et al. A propos d’unrécent foyer de morve du cheval en France. Bull Soc Sci Vet Med Comp 67:309–317, 1965.Van der Schaaf, A. Malleus. In:Van der Hoeden, J., ed. Zoonoses. Amsterdam: Elsevier; 1964.Van Goidsenhoven, C., F. Schoenaers. Maladies Infectieuses des Animaux Domestiques.Liège: Desoer; 1960.


146 BACTERIOSESINFECTION CAUSED BY CAPNOCYTOPHAGACANIMORSUS AND C. CYNODEGMIICD-10 A28.8 other specified zoonotic bacterial <strong>diseases</strong>,not elsewhere classified; T14.1 open wound of unspecified body regionSynonyms: Infection caused by DF-2 <strong>and</strong> DF-2–like bacteria.Etiology: Among the bacterial strains sent for identification <strong>to</strong> the US Centers forDisease Control <strong>and</strong> Prevention (CDC), there was a group that was named DF-2(dysgonic fermenter-2). It consisted of small, gram-negative bacilli that grow slowly<strong>and</strong> with difficulty in <strong>common</strong> labora<strong>to</strong>ry media. The first strain was received in1961 <strong>and</strong> the first report on the human disease—a person bitten by two dogs—waspublished in 1976. Another group was named DF-2–like. These organisms were ultimatelydescribed according <strong>to</strong> the rules of nomenclature <strong>and</strong> bacterial classification.There are two different species: Capnocy<strong>to</strong>phaga canimorsus <strong>and</strong> C. cynodegmi(Brenner et al., 1989). Both species consist of gram-negative bacilli 1 <strong>to</strong> 3 micronslong that form filaments <strong>and</strong> are longer in blood agar. They do not have flagella, butdo have gliding motility. They are microaerophilic <strong>and</strong> grow better in an atmosphere<strong>to</strong> which 5% <strong>to</strong> 10% carbon dioxide has been added. The best medium for theirgrowth is heart infusion agar with 5% sheep or rabbit blood. They are oxidase- <strong>and</strong>catalase-positive, unlike CDC group DF-1 (C. ochracea, C. gingivalis, <strong>and</strong> C. sputigena),which is involved in dental processes <strong>and</strong> is not of zoonotic interest. C. cynodegmidiffers from C. canimorsus in that it ferments raffinose, sucrose, <strong>and</strong> melibiose(Brenner et al., 1989), <strong>and</strong> exhibits marked pathogenic differences.Geographic Distribution: Worldwide, as are their reservoirs <strong>and</strong> sources ofinfection, cats <strong>and</strong> dogs. CDC received strains of C. canimorsus not only from theUS, but also from Australia, Canada, Denmark, France, Great Britain, theNetherl<strong>and</strong>s, New Zeal<strong>and</strong>, South Africa, <strong>and</strong> Sweden.Occurrence in Man: From 1961 <strong>to</strong> February of 1993, CDC received 200 culturesof C. canimorsus isolated from man (CDC, 1993). C. canimorsus occurs primarilyin people who have had a splenec<strong>to</strong>my, alcoholics, <strong>and</strong> those with chronic pulmonarydisease or a malignant blood disease. The disease may occur at any age, butpeople over age 50 predominated in a series of cases. In 77% of the cases, the diseasewas preceded by a dog bite or, less frequently, a cat bite, or some other exposure<strong>to</strong> these animals (a scratch, for example).C. cynodegmi occurs in healthy individuals, without any preceding or concurrentdisease.Occurrence in Animals: C. canimorsus <strong>and</strong> C. cynodegmi have been isolatedfrom the saliva of healthy dogs <strong>and</strong> cats, <strong>and</strong> thus are assumed <strong>to</strong> make up part ofthe normal flora in the mouths of these animals.The Disease in Man: In infections caused by C. canimorsus, the spectrum ofclinical manifestations varies from cellulitis that heals spontaneously <strong>to</strong> fatal septicemia.Serious cases are usually associated with people who have had a splenec<strong>to</strong>myor whose liver has been affected by alcoholism. This would indicate that C.canimorsus is opportunistic <strong>and</strong> not very virulent. However, a fatal case was


CAPNOCYTOPHAGA CANIMORSUS AND C. CYNODEGMI 147described in Australia of a 66-year-old woman with septicemia who was hospitalized48 hours after having been bitten by her dog. The patient presented with symp<strong>to</strong>msof septicemic shock, hemorrhagic eruption, <strong>and</strong> altered consciousness. She hadno prior illness that could have predisposed her <strong>to</strong> this syndrome. She died 16 hoursafter being admitted, despite having received intravenous antibiotic treatment(Clarke et al., 1992).A similar case occurred in Belgium in a 47-year-old woman without any his<strong>to</strong>ryof prior illness. She was admitted <strong>to</strong> the emergency room with septic shock five daysafter receiving a small lesion on the h<strong>and</strong> from her dog. C. canimorsus was isolatedfrom her blood. Despite intensive treatment, she developed multiple organic deficiencies<strong>and</strong> died 27 days after being admitted (Hantson et al., 1991).The clinical picture includes meningitis, endocarditis, septic arthritis, gangrene,disseminated intravascular coagulation, <strong>and</strong> keratitis. The literature records a <strong>to</strong>tal offive cases of ophthalmic infections due <strong>to</strong> cat scratches or close exposure <strong>to</strong> thisanimal. There was also one case attributed <strong>to</strong> a dog (Pa<strong>to</strong>n et al., 1988).Capnocy<strong>to</strong>phaga cynodegmi causes infection in wounds inflicted by dogs. It doesnot produce systemic infection.C. canimorsus <strong>and</strong> C. cynodegmi are sensitive <strong>to</strong> various antibiotics, includingpenicillin, erythromycin, minocycline, <strong>and</strong> doxycycline. Penicillin G is usually preferredby doc<strong>to</strong>rs for wounds caused by dogs (Hicklin et al., 1987). It should be keptin mind that 3% <strong>to</strong> 23% of the gram-negative bacteria isolated from the oropharynxof dogs may be resistant <strong>to</strong> penicillin (Hsu <strong>and</strong> Finberg, 1989).The Disease in Animals: C. canimorsus <strong>and</strong> C. cynodegmi are normal componentsof the bacterial flora in the oropharynx of dogs, cats, sheep, <strong>and</strong> cattle. Theyare not pathogenic for these animal species.Source of Infection <strong>and</strong> Mode of Transmission: The reservoir of the infectionis dogs <strong>and</strong> cats. The source is the saliva of these animals <strong>and</strong> transmission iseffected by a bite.C. canimorsus was isolated from the nose <strong>and</strong> mouth of 4 out of 50 clinically normaldogs (8%). The agent was also isolated from dogs <strong>and</strong> cats whose bites causedinfection in man (Bailie et al., 1978; Chen <strong>and</strong> Fonseca, 1986; Mar<strong>to</strong>ne et al., 1980;Carpenter et al., 1987). A broader study indicated that in a sample of 180 dogs, 24%were carriers of C. canimorsus <strong>and</strong> 11% were carriers of C. cynodegmi; in a sampleof 249 cats, 17% carried C. canimorsus <strong>and</strong> 8% carried C. cynodegmi in theirmouths. The agent was also isolated in a significant percentage of sheep <strong>and</strong> cattle(25% <strong>and</strong> 33%, respectively). In contrast, these agents could not be isolated from thenormal flora of man (Westwell et al., 1989).C. canimorsus is primarily an opportunistic pathogen that infects individualsweakened by concurrent <strong>diseases</strong>. Those who have had a splenec<strong>to</strong>my comprise ahigh-risk group. Asplenic individuals suffer deficient IgM <strong>and</strong> IgG production <strong>and</strong>delayed macrophage mobilization. They also produce less tuftsin, a protein derivedfrom IgG that stimulates phagocy<strong>to</strong>sis (August, 1988). Liver disease caused by alcoholismis another predisposing fac<strong>to</strong>r for the infection. Predisposition is associatedwith susceptibility <strong>to</strong> bacteremia (Kanagasundaram <strong>and</strong> Levy, 1979).Role of Animals in the Epidemiology of the Disease: This is a zoonosis inwhich dogs <strong>and</strong>, <strong>to</strong> a lesser extent, cats, play an essential role.


148 BACTERIOSESDiagnosis: C. canimorsus can be isolated from blood (see the culture medium <strong>and</strong>atmosphere indicated in the section on etiology). In asplenic patients, it is useful <strong>to</strong>make a gram-stained preparation of the leukocyte layer of the extracted blood sample.C. cynodegmi is isolated from wounds.Prevention <strong>and</strong> Control: The treatment for any bite should first be thorough irrigationwith water, then cleaning with soap <strong>and</strong> water. In the case of asplenic patients<strong>and</strong> alcoholics, it is advisable <strong>to</strong> administer antibiotics prophylactically. It is recommendedthat such people not own dogs or cats. However, not all authors agree withthis recommendation.BibliographyAugust, J.R. Dysgonic fermenter-2 infections. J Am Vet Med Assoc 193:1506–1508, 1988.Bailie, W.E., E.C. S<strong>to</strong>we, A.M. Schmitt. Aerobic bacterial flora of oral <strong>and</strong> nasal fluids ofcanines with reference <strong>to</strong> bacteria associated with bites. J Clin Microbiol 7:223–231, 1978.Cited in: August, J.R. Dysgonic fermenter-2 infections. J Am Vet Med Assoc 193:1506–1508,1988.Brenner, D.J., D.G. Hollis, G.R. Fanning, R.E. Weaver. Capnocy<strong>to</strong>phaga canimorsus sp.nov. (formerly CDC group DF-2), a cause of septicemia following dog bite, <strong>and</strong> C. cynodegmisp. nov., a cause of localized wound infection following dog bite. J Clin Microbiol27:231–235, 1989.Carpenter, P.D., B.T. Heppner, J.W. Gnann. DF-2 bacteremia following cat bites. Report oftwo cases. Am J Med 82:621–623, 1987.Chan, P.C., K. Fonseca. Septicemia <strong>and</strong> meningitis caused by dysgonic fermenter-2 (DF-2). J Clin Pathol 39:1021–1024, 1986.Clarke, K., D. Devonshire, A. Veitch, et al. Dog-bite induced Capnocy<strong>to</strong>phaga canimorsussepticemia. Aust New Zeal<strong>and</strong> J Med 22:86–87, 1992.Hantson, P., P.E. Gautier, M.C. Vekemans, et al. Fatal Capnocy<strong>to</strong>phaga canimorsus septicemiain a previously healthy woman. Ann Emerg Med 20:93–94, 1991.Hicklin, H., A. Verghese, S. Alvarez. Dysgonic fermenter 2 septicemia. Rev Infect Dis9:884–890, 1987.Hsu, H-W., R.W. Finberg. Infections associated with animal exposure in two infants. RevInfect Dis 11:108–115, 1989.Kanagasundaram, N., C.M. Levy. Immunologic aspects of liver disease. Med Clin NorthAm 63:631–642, 1979. Cited in: Hicklin, H., A. Verghese, S. Alvarez. Dysgonic fermenter 2septicemia. Rev Infect Dis 9:884–890, 1987.Mar<strong>to</strong>ne, W.J., R.W. Zuehl, G.E. Minson, W.M. Scheld. Postsplenec<strong>to</strong>my sepsis with DF-2: Report of a case with isolation of the organism from the patient’s dog. Ann Intern Med93:457–458, 1980. Cited in: August, J.R. Dysgonic fermenter-2 infections. J Am Vet MedAssoc 193:1506–1508, 1988.Pa<strong>to</strong>n, B.G., L.D. Ormerod, J. Peppe, K.R. Kenyon. Evidence for a feline reservoir of dysgonicfermenter 2 keratitis. J Clin Microbiol 26:2439–2440, 1988.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Capnocy<strong>to</strong>phaga canimorsus sepsis misdiagnosed asplague—New Mexico, 1992. MMWR Morb Mortal Wkly Rep 42:72–73, 1993.Westwell, A.J., K. Kerr, M.B. Spencer, et al. DF-2 infection. Br Med J 298:116–117, 1989.


LEPROSY 149LEPROSYICD-10 A30.9 leprosy, unspecifiedSynonyms: Hansen’s disease, hanseniasis.Etiology: Mycobacterium leprae,a polymorphic acid-alcohol-fast bacillus that up<strong>to</strong> now has been impossible <strong>to</strong> culture in artificial labora<strong>to</strong>ry media or in tissue cultures.M. leprae is difficult <strong>to</strong> distinguish from other unculturable mycobacteria naturallyaffecting animals.The failure of attempts <strong>to</strong> culture M. leprae in vitro constitutes a great barrier <strong>to</strong>better determining its biochemical characteristics for identification purposes as wellas for therapeutic <strong>and</strong> immunologic studies. In part, this difficulty has been overcome,first by in vivo culture on mouse footpads <strong>and</strong> later in nine-b<strong>and</strong>ed armadillos(Dasypus novemcinctus). At present, the latter serve as a model for leproma<strong>to</strong>usleprosy <strong>and</strong> provide a large number of bacilli for research.In identification of M. leprae, the dopa (3,4-dihydroxyphenylalanine) oxidationtest <strong>and</strong> extraction with pyridine are of value. Homogenate of human leproma (granuloma<strong>to</strong>usnodule rich in M. leprae <strong>and</strong> characteristic of leproma<strong>to</strong>us leprosy) oxidizesdopa <strong>to</strong> indole. Extraction with pyridine eliminates the acid-fast quality of M.leprae, but not that of other mycobacteria.In recent years, more precise identification of M. leprae has been achieved bystructural analysis of its mycolic acids, analysis by immunodiffusion of its antigens,<strong>and</strong> interaction of leprosy bacilli with bacteriophages specific for mycobacteria(Ras<strong>to</strong>gi et al., 1982).Occurrence in Man: Leprosy is endemic in 93 countries. Eighty percent of allrecorded cases are concentrated in five countries: India, Brazil, Nigeria, Myanmar(Burma), <strong>and</strong> Indonesia (<strong>WHO</strong>, 1988). The highest prevalence is found in the tropical<strong>and</strong> subtropical regions of Asia, Africa, Latin America, <strong>and</strong> Oceania. Leprosy isvery prevalent in India, Southeast Asia, the Philippines, Korea, southern China,Papua New Guinea, <strong>and</strong> some Pacific isl<strong>and</strong>s. Ninety percent of the cases reported inLatin America come from five countries: Argentina, Brazil, Colombia, Mexico, <strong>and</strong>Venezuela (Brubaker, 1983). Chile is the only South American country free of theinfection. In the United States, most cases occur among immigrants. Au<strong>to</strong>chthonouscases arise in Hawaii, Puer<strong>to</strong> Rico, Texas, <strong>and</strong> Louisiana. The infection’s prevalenceis related <strong>to</strong> the population’s socioeconomic level. The fact that the disease has practicallydisappeared in Europe is attributed <strong>to</strong> the improved st<strong>and</strong>ard of living there.There are differences in the regional or racial prevalence of tuberculoid <strong>and</strong> leproma<strong>to</strong>usleprosy. Ninety percent of the cases in endemic areas of Africa <strong>and</strong> 80%of the cases in India are of the tuberculoid type. The leproma<strong>to</strong>us form represents30% <strong>to</strong> 50% of cases among the white population or in some Asian countries suchas Japan, China, <strong>and</strong> Korea (Bechelli et al., 1972).In countries with efficient control programs, it was expected that prevalencewould fall by 60% <strong>to</strong> 80% by the year 2000. Of the cases reported worldwide, 49.1%were under multidrug treatment in 1990 (Noorden, 1990). The cumulative rate ofcoverage with polychemotherapy has reached 82%. Each year 1.4 million patientsare freed from the disease (<strong>WHO</strong>, 1993).


150 BACTERIOSESOccurrence in Animals: Natural infection has been found in nine-b<strong>and</strong>edarmadillos (Dasypus novemcinctus) in Louisiana <strong>and</strong> Texas (USA) <strong>and</strong> in Mexico.By 1983, the infection had been observed in some 100 armadillos captured inLouisiana (Meyers et al., 1983). Depending on their place of origin, between 4%<strong>and</strong> 29.6% of 1,033 armadillos examined were infected. On the Gulf Coast of Texas,leprosy lesions were found in 4.66% of 451 armadillos captured (Smith et al., 1983).The disease form found in these animals was a leproma<strong>to</strong>us leprosy identical <strong>to</strong> thetype produced by experimental inoculation with material from humans. On the otherh<strong>and</strong>, the search for naturally infected armadillos carried out by other researchers inLouisiana, Texas, <strong>and</strong> Florida, as well as in Colombia <strong>and</strong> Paraguay, produced negativeresults (Kirchheimer, 1979).At present, natural infection of nine-b<strong>and</strong>ed armadillos is a well-established fact.Its distribution is limited <strong>to</strong> some states in the US <strong>and</strong> Mexico. In Mexico, 1 of 96armadillos was positive based on his<strong>to</strong>pathology <strong>and</strong> mouse footpad inoculation(Amezcua et al., 1984). In a study of armadillos found dead on the highways ofLouisiana, 10 of 494 (2%) were positive based on his<strong>to</strong>pathology <strong>and</strong> pyridineextraction (Job et al., 1986a). The infection was also confirmed in an armadillo atthe San Diego Zoo, California, <strong>and</strong> in another armadillo at the Centers for DiseaseControl <strong>and</strong> Prevention. The two armadillos originally came from Texas (Walsh etal., 1981).The ELISA method was adopted for serological study of leprosy in armadillosusing phenolic glycolipid (PGL-1) antigen (Truman et al., 1986), which is consideredspecific for M. leprae (Young <strong>and</strong> Buchanan, 1983). This test was conductedon armadillos captured in central Louisiana before being used in the labora<strong>to</strong>ry(1960–1964). It was found that 17 of the 182 sera (9.3%) were serologically positive.This study was undertaken <strong>to</strong> refute the argument that free armadillos couldhave become infected by experimental armadillos through carelessness. The serawere collected at that time for a study on lep<strong>to</strong>spirosis. Of 20 armadillos capturedshortly before this study, four were positive.Another study used ELISA <strong>and</strong> his<strong>to</strong>pathological tests <strong>to</strong> examine 77 armadillosin an estimated population of 254 ± 60 animals in a defined area of Louisiana. Fiveof 67 (1.5%) sera tested with ELISA <strong>and</strong> 1 of 74 (1.3%) ears submitted forhis<strong>to</strong>pathological examination were positive (Stallknecht et al., 1987).On the Texas Gulf Coast, the presence of leprosy in armadillos was demonstrated(Smith et al., 1983). More recently, 237 armadillo ears from 51 central Texas districtswere examined; no positives were found upon his<strong>to</strong>logical examination (Clarket al., 1987). A similar negative result was obtained for 853 ears from armadilloskilled on the highways or captured for research purposes in five southeastern USstates. The examination included microscopic <strong>and</strong> his<strong>to</strong>pathological examination(Howerth et al., 1990). An infected animal had previously been found in the state ofMississippi (Walsh et al., 1986).A spontaneous case of leprosy similar <strong>to</strong> the borderline or dimorphous form wasdescribed in a chimpanzee imported from Sierra Leone <strong>to</strong> the United States. Clinical<strong>and</strong> his<strong>to</strong>pathologic characteristics (with invasion of dermal nerves by the agent)were identical <strong>to</strong> those of the human disease. Attempts <strong>to</strong> culture the bacteria werenegative, <strong>and</strong> the chimpanzee did not respond <strong>to</strong> tuberculin or lepromin, just ashumans infected with leproma<strong>to</strong>us or dimorphous leprosy give a negative reaction.As with M. leprae of human origin, experimental inoculation of rats with the iso-


LEPROSY 151lated bacillus produced neither disease nor lesions. The only differences with M.leprae of human origin were negative results <strong>to</strong> the dopa oxidation <strong>and</strong> pyridinetests. However, the dopa oxidation test sometimes fails in animals (armadillos) inoculatedexperimentally with human M. leprae (Donham <strong>and</strong> Leininger, 1977).Results obtained by inoculating mouse foot pads were similar <strong>to</strong> those derived withM. leprae of human origin, i.e., reproduction of the bacterium in six months up <strong>to</strong> aquantity similar <strong>to</strong> that of M. leprae without dissemination from the inoculationpoint (Leininger et al., 1978).Another case of naturally acquired leprosy was discovered in a primate,Cercocebus atys or sooty mangabey monkey (identified in one publication asCercocebus <strong>to</strong>rquatus atys), captured in West Africa <strong>and</strong> imported <strong>to</strong> the UnitedStates in 1975 (Meyers et al., 1980, 1981). The clinical picture <strong>and</strong> his<strong>to</strong>pathologywere similar <strong>to</strong> man’s <strong>and</strong> the etiologic agent was identified as M. leprae based onthe following criteria: invasion of the host’s nerves, staining properties, electronmicroscopy findings, inability <strong>to</strong> grow in mycobacteriologic media, positive dopaoxidation reaction, reactivity <strong>to</strong> lepromin, patterns of infection in mice <strong>and</strong> armadillos,sensitivity <strong>to</strong> sulfones, <strong>and</strong> DNA homology (Meyers et al.,1985). Simultaneousintravenous <strong>and</strong> intracutaneous inoculation succeeded in reproducing the infection<strong>and</strong> disease in other Cercocebus monkeys. The early appearance of signs (5 <strong>to</strong> 14months), varying clinical disease forms, neuropathic deformities, bacillemia, <strong>and</strong>dissemination <strong>to</strong> various cool parts of the body make the mangabey monkey potentiallythe most complete model for the study of leprosy. It is the third animal speciesreported <strong>to</strong> be able <strong>to</strong> acquire leprosy by natural infection (Walsh et al., 1981;Meyers et al., 1983, 1985).The Disease in Man: The incubation period is usually 3 <strong>to</strong> 5 years, but it can varyfrom 6 months <strong>to</strong> 10 years or more (Bullock, 1982). Clinical forms of leprosy covera wide spectrum, ranking from mild self-healing lesions <strong>to</strong> a progressive <strong>and</strong> destructivechronic disease. Tuberculoid leprosy is found at one end of the spectrum <strong>and</strong> leproma<strong>to</strong>usleprosy at the other. Between them are found the intermediate forms.Tuberculoid leprosy is characterized by often asymp<strong>to</strong>matic localized lesions ofthe skin <strong>and</strong> nerves. Basically, the lesion consists of a granuloma<strong>to</strong>us, paucibacillary,inflamma<strong>to</strong>ry process. The bacilli are difficult <strong>to</strong> detect, <strong>and</strong> can be observedmost frequently in the nerve endings of the skin. This form results from activedestruction of the bacilli by the undeteriorated cellular immunity of the patient. Onthe other h<strong>and</strong>, the humoral response generally involves low titers. Nerve destructioncauses lowered conduction; heat sensibility is the most affected, tactile sensibilityless so. Trophic <strong>and</strong> au<strong>to</strong>nomic changes are <strong>common</strong>, especially ulcers on thesole <strong>and</strong> mutilation of limbs (Toro-González et al., 1983).Leproma<strong>to</strong>us leprosy is characterized by numerous symmetrical skin lesions consistingof macules <strong>and</strong> diffuse infiltrations, plaques, <strong>and</strong> nodules of varying sizes(lepromas). There is involvement of the mucosa of the upper respira<strong>to</strong>ry tract, oflymph nodes, liver, spleen, <strong>and</strong> testicles. Infiltrates are basically histiocytes with afew lymphocytes. Cellular immunity is absent (negative reaction <strong>to</strong> lepromin) <strong>and</strong>antibody titers are high. In this form of the disease, as in the dimorphous, erythemanodosum leprosum (ENL) often appears.The indeterminate form of leprosy has still not been adequately defined from theclinical st<strong>and</strong>point; it is considered <strong>to</strong> be the initial state of the disease. The first


152 BACTERIOSEScutaneous lesions are flat, hypopigmented, <strong>and</strong> have ill-defined borders. If this initialform is not treated, it may develop in<strong>to</strong> tuberculoid, dimorphous, or leproma<strong>to</strong>usleprosy. Bacilli are few <strong>and</strong> it is difficult <strong>to</strong> confirm their presence.Finally, the dimorphous or borderline form occupies an intermediate positionbetween the two polar forms (tuberculoid <strong>and</strong> leproma<strong>to</strong>us), <strong>and</strong> shares propertiesof both; it is unstable <strong>and</strong> may progress in either direction. Destruction of nervetrunks may be extensive. Bacilli are observed in scrapings taken from skin lesions.A study group (<strong>WHO</strong>, 1985) has, primarily for practical treatment purposes,defined two types of the disease.“a) Paucibacillary: This includes the categories described as indeterminate (I) <strong>and</strong>tuberculoid (T) leprosy in the Madrid classification, <strong>and</strong> the indeterminate (I), polartuberculoid (TT) <strong>and</strong> borderline tuberculoid (BT) categories in the Ridley <strong>and</strong>Jopling classification, whether diagnosed clinically or his<strong>to</strong>pathologically with abacterial index of


LEPROSY 153were first observed, the animal began <strong>to</strong> suffer deformities <strong>and</strong> paralysis of theextremities. His<strong>to</strong>pathologic findings indicated the subpolar or intermediate leproma<strong>to</strong>usform, according <strong>to</strong> the Ridley <strong>and</strong> Jopling classification. The disease wasprogressive, with neuropathic deformation of the feet <strong>and</strong> h<strong>and</strong>s. It seemed <strong>to</strong>regress when specific treatment was administered. The animal apparently acquiredthe disease from a patient with active leprosy. Experimental infections carried out <strong>to</strong>date have indicated that these animals may experience a spectrum of different formssimilar <strong>to</strong> those seen in man (Meyers et al., 1985).Source of Infection <strong>and</strong> Mode of Transmission: Man is the principal reservoirof M. leprae. The method of transmission is still not well known due <strong>to</strong> the extendedincubation period. Nevertheless, the principal source of infection is believed <strong>to</strong> beleproma<strong>to</strong>us patients, in whom the infection is multibacillary, skin lesions are oftenulcerous, <strong>and</strong> a great number of bacilli are shed through the nose; similarly, bacilliare found in the mouth <strong>and</strong> pharynx. Consequently, transmission might be broughtabout by contact with infected skin, especially if there are abrasions or wounds.Currently, particular importance is attributed <strong>to</strong> aerosol transmission. Nasal secretionsfrom leproma<strong>to</strong>us patients contain approximately 100 million bacilli per milliliter.In addition, M. leprae can survive for about seven days in dried secretions.Another possible route of transmission is mother’s milk, which contains a largenumber of bacilli in leproma<strong>to</strong>us patients (Bullock, 1990). Oral transmission <strong>and</strong>transmission by hema<strong>to</strong>phagous arthropods are not discounted, but they are assignedless epidemiological importance.Until recently, leprosy was believed <strong>to</strong> be an exclusively human disease. However,research in recent years has demonstrated that the infection <strong>and</strong> the disease alsooccur naturally in wild animals. Although some researchers (Kirchheimer, 1979)have expressed doubt that the animal infection is identical <strong>to</strong> the human, the accumulatedevidence indicates that the etiologic agent is the same. The criteria (Binfordet al., 1982) used <strong>to</strong> identify the bacillus in animals as M. leprae were as follows:(1) selective invasion of the peripheral nerves by bacilli, since the onlyMycobacterium known <strong>to</strong> date <strong>to</strong> invade the nerves is M. leprae, (2) failure <strong>to</strong> growon <strong>common</strong> labora<strong>to</strong>ry media for mycobacteria, (3) positive pyridine test <strong>to</strong> eliminateacid-fastness, (4) positive dopa test, (5) characteristic multiplication in mousefoot pads <strong>and</strong> in armadillos, <strong>and</strong> (6) reactivity of lepromin prepared with animalbacilli compared <strong>to</strong> that of st<strong>and</strong>ard lepromin.The origin of the infection in animals is unknown. Some authors believe thatarmadillos contracted the infection from a human source, perhaps from multibacillarypatients before the era of sulfones. In this regard, it should be pointed out thatleprosy bacilli may remain viable for a week in dried nasal secretions <strong>and</strong> thatarmadillos are in close contact with the soil. The high prevalence in some localitieswould also indicate that armadillos can transmit the disease <strong>to</strong> each other, either byinhalation or direct contact. Another possible transmission vehicle is maternal milk,in which the agent has been detected (Walsh et al., 1981). It has also been suggestedthat transmission among armadillos may be brought about by thorns penetrating theears, nose, or other body parts (Job et al., 1986b), as apparently armadillos useplaces with spiny plants <strong>to</strong> hide from their preda<strong>to</strong>rs. These authors have foundthorns in the ears of 25.5% of 494 armadillos captured in Louisiana, <strong>and</strong> in the noseof 36.6% of them.


154 BACTERIOSESIt is difficult <strong>to</strong> demonstrate that armadillos are a source of infection for manbecause of the long incubation period <strong>and</strong> the impossibility of excluding a humansource in an endemic area. In Texas, a case of human leprosy was attributed <strong>to</strong> apatient’s practice of capturing armadillos <strong>and</strong> eating their meat (Freiberger <strong>and</strong>Fudenberg, 1981). Subsequently, another five cases with h<strong>and</strong> lesions were detectedin natives of the same state who habitually hunted <strong>and</strong> cleaned armadillos but hadno known contact with leprosy patients (Lumpkin III et al., 1983). To determine ifthere was a significant association between contact with armadillos <strong>and</strong> human leprosyin Louisiana, a group of 19 patients was compared with another group of 19healthy individuals from the same area. Of those with leprosy, four had had contactwith armadillos, as opposed <strong>to</strong> five in the control group. Consequently, it was concludedthat such an association did not exist (Filice et al., 1977). However, this conclusionwas questioned, since the only valid comparison would be between personswho have h<strong>and</strong>led armadillos <strong>and</strong> those who have had no contact with them(Lumpkin III et al., 1983).The prevalence of leprosy in armadillos in Louisiana <strong>and</strong> Texas suggests thatthese animals could serve as a reservoir of M. leprae. However, nothing is knownabout the frequency of infection in nonhuman primates <strong>and</strong> the role they may playin transmission of the disease. The sources of the cases of leprosy in these animalswere probably people with leproma<strong>to</strong>us leprosy.Diagnosis: Clinically, an anesthetic or hypoesthetic cutaneous lesion raises suspicionof leprosy, even more so if the nerves are enlarged. Diagnosis is confirmed bybiopsy of the skin lesion, which also permits classification of the form of the leprosy.For patients with leproma<strong>to</strong>us or borderline leproma<strong>to</strong>us leprosy, diagnosis can bemade by using the Ziehl-Neelsen staining technique on a film of nasal mucosa scrapingsor the interphase between erythrocytes <strong>and</strong> leukocytes from a centrifuged bloodsample. His<strong>to</strong>pathologic preparations do not stain well using Ziehl-Neelsen <strong>and</strong> consequentlya Fite-Faraco stain is recommended. Also used is the simplified stainingmethod consisting of eliminating acid-fastness with pyridine in order <strong>to</strong> differentiateM. leprae (Convit <strong>and</strong> Pinardi, 1972). In tuberculoid <strong>and</strong> other paucibacillary formsof leprosy, it is difficult <strong>and</strong> at times impossible <strong>to</strong> confirm the presence of the etiologicagent; in any case, examination of many his<strong>to</strong>logic sections is recommended inorder <strong>to</strong> detect any bacteria present, especially in the nerve endings.Skin tests have no diagnostic value, but they do serve as an aid <strong>to</strong> prognosis.Patients with tuberculoid leprosy or other paucibacillary forms react positively <strong>to</strong>the intradermal lepromin or Mitsuda test (with dead M. leprae bacilli <strong>and</strong> a readingafter 28 days), since their cellular immunity is generally not affected. In contrast,leproma<strong>to</strong>us leprosy <strong>and</strong> other multibacillary forms give negative results <strong>to</strong> theMitsuda test. The lepromin test has limited value for detecting infection in those incontact with patients or the general population in an endemic area (Jacobson, 1991).Serological tests are also of limited use.The ELISA technique (Young <strong>and</strong> Buchanan, 1983) for measuring PGL-1 (phenolicglycolipid antigen) antibodies is a great step forward. The reactive titerdepends on the patient’s bacillary load <strong>and</strong> also serves <strong>to</strong> detect infection in thosewho are in contact with multibacillary patients, as well as in some people in endemicareas (Jacobson, 1991). In Malawi, Africa, where most cases are paucibacillary, thetest was not sufficiently sensitive (unless its specificity were <strong>to</strong> be sacrificed), but it


LEPROSY 155was used <strong>to</strong> detect a high percentage of multibacillary patients (Burgess et al.,1988). A variation of this test is the use of the synthetic disaccharide epi<strong>to</strong>pe ofPGL-1 as an antigen (Brett et al., 1986).Control: Control is based on early detection <strong>and</strong> chemotherapy. Given the multipleconfirmed cases of resistance <strong>to</strong> dapsone, combination of this medication withrifampicin is presently recommended for paucibacillary leprosy, <strong>and</strong> the same twomedications in combination with clofazimine are recommended for multibacillaryleprosy. Rifampicin has a rapid bactericidal effect <strong>and</strong> eliminates contagion inpatients in one <strong>to</strong> two weeks. To achieve the objective of eliminating leprosy, allpatients should receive polychemotherapy. This treatment has been successful inreducing general prevalence from 5.4 million in 1986 <strong>to</strong> 3.7 million in 1990.Widespread testing began in 1992 on a new oral treatment that was developed overthe preceding five years <strong>and</strong> combines two antibiotics, rifampicin <strong>and</strong> ofloxacin.Ofloxacin inhibits an enzyme that controls the way that DNA coils inside the bacterium.It is hoped that this combination will be able <strong>to</strong> cure leprosy in the course ofone month. If testing is successful, all patients should have access <strong>to</strong> this medication(<strong>WHO</strong>, 1992). The isolation of patients in leprosariums is no longer necessary, sincemedication is effective in suppressing infectiousness <strong>and</strong> thus interrupts transmissionof the disease.BibliographyAmezcua, M.E., A. Escobar-Gutierrez, E.E. S<strong>to</strong>rrs, et al. Wild Mexican armadillo with leprosy-likeinfection [letter]. Int J Lepr Other Mycobact Dis 52:254–255, 1984.Bechelli, L.M., V. Martínez Domínguez. Further information on the leprosy problem in theworld. Bull World Health Organ 46:523–536, 1972. Cited in: Bullock, W.E. Mycobacteriumleprae (Leprosy). In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E. Bennett, eds. Principles <strong>and</strong>Practice of Infectious Diseases. 3rd ed. New York: Churchill Livings<strong>to</strong>ne, Inc.; 1990.Binford, C.H., W.M. Meyers, G.P. Walsh. Leprosy. JAMA 247:2283–2292, 1982.Binford, C.H., W.M. Meyers, G.P. Walsh, E.E. S<strong>to</strong>rrs, H.L. Brown. Naturally acquired leprosy-likedisease in the nine-b<strong>and</strong>ed armadillo (Dasypus novemcinctus): His<strong>to</strong>pathologic <strong>and</strong>microbiologic studies of tissues. J Reticuloendothel Soc 22:377–388, 1977.Brett, S.J., S.N. Payne, J. Gigg, et al. Use of synthetic glycoconjugates containing theMycobacterium leprae specific <strong>and</strong> immunodominant epi<strong>to</strong>pe of phenolic glycolipid I in theserology of leprosy. Clin Experim Immunol 64:476–483, 1986.Brubaker, M. Leprosy Control in the Americas. Part I: General Considerations. In: Bolivar’sBicentennial Seminar on Leprosy Control: Report: Caracas 12–14 September 1983. PanAmerican Health Organization, 1983. (PNSP/84–05).Bullock, W.E. Leprosy (Hansen’s disease). In:Wyngaarden, J.B., L.H. Smith, Jr., eds. CecilTextbook of Medicine. 16th ed. Philadelphia: W.B. Saunders; 1982.Bullock, W.E. Mycobacterium leprae (Leprosy). In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E.Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: ChurchillLivings<strong>to</strong>ne, Inc.; 1990.Burgess, P.J., P.E. Fine, J.M. Ponnighaus, C. Draper. Serological tests in leprosy. Thesensitivity, specificity <strong>and</strong> predictive value of ELISA tests based on phenolic glycolipid antigens,<strong>and</strong> the implications for their use in epidemiological studies. Epidemiol Infect101:159–171, 1988.Clark, K.A., S.H. Kim, L.F. Boening, et al. Leprosy in armadillos (Dasypus novemcinctus)from Texas. J Wildl Dis 220–224, 1987.


156 BACTERIOSESConvit, J., M.E. Pinardi. A simple method for the differentiation of Mycobacterium lepraefrom other mycobacteria through routine staining technics. Int J Lepr Other Mycobact Dis40:130–132, 1972.Donham, K.J., J.R. Leininger. Spontaneous leprosy-like disease in a chimpanzee. J InfectDis 136:132–136, 1977.Filice, G.A., R.N. Greenberg, D.W. Fraser. Lack of observed association between armadillocontact <strong>and</strong> leprosy in humans. Am J Trop Med Hyg 26:137–139, 1977.Fine, P.E. Leprosy: The epidemiology of a slow bacterium. Epidemiol Rev 4:161–188, 1982.Freiberger, H.F., H.H. Fudenberg. An appetite for armadillo. Hosp Practice 16:137–144, 1981.Howerth, E.W., D.E. Stallknecht, W.R. Davidson, E.J. Wentworth. Survey for leprosy innine-b<strong>and</strong>ed armadillos (Dasypus novemcinctus) from the southeastern United States. J WildlDis 26:112–115, 1990.Jacobson, R.R. Leprosy. In: Evans, A.S., P.S. Brachman, eds. Bacterial Infections ofHumans. 2nd ed. New York: Plenum; 1991.Job, C.K., E.B. Harris, J.L. Allen, R.C. Hastings. A r<strong>and</strong>om survey of leprosy in wild nineb<strong>and</strong>edarmadillos in Louisiana. Int J Lepr Other Mycobact Dis 54:453–457, 1986a.Job, C.K., E.B. Harris, J.L. Allen, R.C. Hastings. Thorns in armadillo ears <strong>and</strong> noses <strong>and</strong>their role in the transmission of leprosy. Arch Pathol Lab Med 110:1025–1028, 1986b.Job, C.K., R.M. Sánchez, R.C. Hastings. Manifestations of experimental leprosy in thearmadillo. Am J Trop Med Hyg 34:151–161, 1985.Kirchheimer, W.F. Leprosy (Hansen’s Disease). In: S<strong>to</strong>enner, H., W. Kaplan, M. Torten, eds.Vol I, Section A: CRC H<strong>and</strong>book Series in Zoonoses. Boca Ra<strong>to</strong>n: CRC Press; 1979.Kirchheimer, W.F., E.E. S<strong>to</strong>rrs, C.H. Binford. Attempts <strong>to</strong> establish the armadillo (Dasypusnovemcinctus Linn) as model for the study of leprosy. II. His<strong>to</strong>pathologic <strong>and</strong> bacteriologicpost-mortem findings in leproma<strong>to</strong>id leprosy in the armadillo. Int J Lepr Other Mycobact Dis40:229–242, 1972.Leininger, J.R., K.J. Donham, W.M. Meyers. Leprosy in a chimpanzee. Postmortem lesions.Int J Lepr Other Mycobact Dis 48:414–421, 1980.Leininger, J.R., K.J. Donham, M.J. Rubino. Leprosy in a chimpanzee. Morphology of theskin lesions <strong>and</strong> characterization of the organism. Vet Pathol 15:339–346, 1978.Lumpkin III, L.R., G.F. Cox, J.E. Wolf, Jr. Leprosy in five armadillo h<strong>and</strong>lers. J Am AcadDerma<strong>to</strong>l 9:899–903, 1983.Martin, L.N., B.J. Gormus, R.H. Wolf, G.P. Walsh, W.M. Meyers, C.H. Binford, et al.Experimental leprosy in nonhuman primates. Adv Vet Sci Comp Med 28:201–236, 1984.Meyers, W.M., G.P. Walsh, C.H. Binford, H.L. Brown, R.H. Wolf, B.J. Gormus, et al.Multibacillar leprosy in unaltered hosts, with emphasis on armadillos <strong>and</strong> monkeys [abstract].Int J Lepr Other Mycobact Dis 50:584–585, 1982.Meyers, W.M., G.P. Walsh, C.H. Binford, R.H. Wolf, B.J. Gormus, L.N. Martin, et al.Models of multibacillary leprosy in unaltered hosts: Current Status. In: Bolivar’s BicentennialSeminar on Leprosy Control: Report: Caracas 12–14 September 1983. Pan American HealthOrganization, 1983. (PNSP/84–05).Meyers, W.M., G.P. Walsh, H.L. Brown, C.H. Binford, P.J. Gerone, R.H. Wolf, et al. Leprosyin a mangabey monkey (Cercocebus <strong>to</strong>rquatus atys “sooty” mangabey) [abstract]. Int J LeprOther Mycobact Dis 49:500–502, 1981.Meyers, W.M., G.P. Walsh, H.L. Brown, C.H. Binford, G.D. Imes, Jr., T.L. Hadfield, et al.Leprosy in a mangabey monkey—naturally acquired infection. Int J Lepr Other Mycobct Dis53:1–14, 1985.Meyers, W.M., G.P. Walsh, H.L. Brown, Y. Fukunishi, C.H. Binford, P.J. Gerone, et al.Naturally-acquired leprosy in a mangabey monkey (Cercocebus spp.) [summary]. Int J LeprOther Mycobact Dis 48:495–496, 1980.Noorden, S.K. Multidrug therapy (MDT) <strong>and</strong> leprosy control. Indian J Lepr 62:448–458, 1990.


LEPTOSPIROSIS 157Ras<strong>to</strong>gi, N., C. Frehel, A. Ryter, H.L. David. Comparative ultrastructure of Mycobacteriumleprae <strong>and</strong> M. avium grown in experimental hosts. Ann Microbiol 133:109–128, 1982.Smith, J.H., D.S. Folse, E.G. Long, J.D. Christie, D.T. Crouse, M.E. Tewes, et al. Leprosyin wild armadillos (Dasypus novemcinctus) of the Texas Gulf Coast: Epidemiology <strong>and</strong>mycobacteriology. J Reticuloendothel Soc 34:75–88, 1983.Stallknecht, D.E., R.W. Truman, M.E. Hugh-Jones, C.K. Job. Surveillance for naturallyacquired leprosy in a nine-b<strong>and</strong>ed armadillo population. J Wildl Dis 23:308–310, 1987.Toro-González, G., G. Román-Campos, L. Navarro de Román. Leprosy: neurología tropical.Bogotá: Printer Colombiana; 1983.Truman, R.W., E.J. Shannon, H.V. Hagstad, et al. Evaluation of the origin of Mycobacteriumleprae infections in the wild armadillo, Dasypus novemcinctus. Am J Trop Med Hyg35:588–593, 1986.Walsh, G.P., W.M. Meyers, C.H. Binford. Naturally acquired leprosy in the nine-b<strong>and</strong>edarmadillo: A decade of experience 1975–1985. J Leukoc Biol 40:645–656, 1986. Cited in:Howerth, E.W., D.E. Stallknecht, W.R. Davidson, E.J. Wentworth. Survey for leprosy in nineb<strong>and</strong>edarmadillos (Dasypus novemcinctus) from the southeastern United States. J Wildl Dis26:112–115, 1990.Walsh, G.P., W.M. Meyers, C.H. Binford, P.J. Gerone, R.H. Wolf, J.R. Leininger. Leprosy—a zoonosis. Lepr Rev 52(Suppl.1):77–83, 1981.Walsh, G.P., E.E. S<strong>to</strong>rrs, W.M. Meyers, C.H. Binford. Naturally acquired leprosy-like diseasein the nine-b<strong>and</strong>ed armadillo (Dasypus novemcinctus): Recent epizootiologic findings. JReticuloendothel Soc 22:363–367, 1977.World Health Organization (<strong>WHO</strong>). Epidemiology of leprosy in relation <strong>to</strong> control. Repor<strong>to</strong>f a <strong>WHO</strong> Study Group. Geneva: <strong>WHO</strong>, 1985. (Technical Report Series 716).World Health Organization (<strong>WHO</strong>). <strong>WHO</strong> Expert Committee on Leprosy, Sixth Report.Geneva: <strong>WHO</strong>; 1988. (Technical Report Series 768).World Health Organization (<strong>WHO</strong>). Trials begin of new treatment for leprosy—diseasecould be conquered [press release]. Geneva: <strong>WHO</strong>; 3 February 1992. (Press Release <strong>WHO</strong> 5).World Health Organization (<strong>WHO</strong>). Progress <strong>to</strong>wards the elimination of leprosy as a publichealth problem. Wkly Epidemiol Rec 68(25):181–186, 1993.Young, D.B., T.M. Buchanan. A serological test for leprosy with a glycolipid specific forMycobacterium leprae. Science 221:1057–1059, 1983.LEPTOSPIROSISICD-10 A27.0 lep<strong>to</strong>spirosis icterohaemorrhagica;A27.8 other forms of lep<strong>to</strong>spirosisSynonyms: Weil’s disease, swineherd’s disease, rice-field fever, cane-cutter’sfever, swamp fever, mud fever, <strong>and</strong> other local names; Stuttgart disease, canicolafever (dogs).Etiology: Lep<strong>to</strong>spires are spiral-shaped bacteria, with open, hooked ends; theyare motile, aerobic, <strong>and</strong> culturable, <strong>and</strong> they measure some 6 <strong>to</strong> 20 microns long by0.1 microns in diameter. They can be seen under a dark-field microscope <strong>and</strong> pass


158 BACTERIOSESthrough filters that block other bacteria. Two species are recognized: Lep<strong>to</strong>spirainterrogans <strong>and</strong> L. biflexa. L. interrogans is pathogenic for man <strong>and</strong> animals; L.biflexa, a free-living saprophyte found in surface waters, is seldom associated withinfection in mammals.The species of interest as a zoonotic agent is L. interrogans. It has more than 200serologic variants, or serovars, which constitutes the basic taxon. Serovars aregrouped for convenience in<strong>to</strong> 23 serogroups (which is not a recognized taxon) on thebasis of the predominant agglutinogenic components they share (Faine, 1982;Alex<strong>and</strong>er, 1991). Through the use of ribosomal RNA gene restriction patterns, anattempt is being made <strong>to</strong> characterize L. interrogans serovars in order <strong>to</strong> establishthe bases for molecular typing (Perolat et al., 1990).Geographic Distribution: Worldwide. There are universal serovars, such as L.interrogans serovar icterohaemorrhagiae <strong>and</strong> serovar canicola, <strong>and</strong> serovars tha<strong>to</strong>ccur only in certain regions. Each region has characteristic serotypes, determinedby its ecology. Lep<strong>to</strong>spirosis has a high prevalence in tropical countries with heavyrainfall <strong>and</strong> neutral or alkaline soils.Occurrence in Man: Incidence varies in different parts of the world. The diseasemay occur sporadically or in epidemic outbreaks. In general, outbreaks are causedby exposure <strong>to</strong> water contaminated by the urine of infected animals. Several occupationalgroups are particularly at risk, such as workers in rice fields, sugarcaneplantations, mines, sewer systems, <strong>and</strong> slaughterhouses, as well as animal caretakers,veterinarians, <strong>and</strong> members of the military.Occurrence in Animals: The infection is <strong>common</strong> in rodents <strong>and</strong> other wild <strong>and</strong>domestic mammals. Worldwide, the infection occurs in approximately 160 mammalianspecies (Alex<strong>and</strong>er, 1991). Each serovar has its preferred animal host orhosts, but each animal species may be host <strong>to</strong> one or more serovars. Thus, for example,the serovar pomona has pigs <strong>and</strong> cattle as its principal hosts, but it may transi<strong>to</strong>rilyinfect other host animals. Dogs are the principal reservoir of canicola, but itmay occasionally be found in foxes, swine, <strong>and</strong> cattle.The Disease in Man: Man is susceptible <strong>to</strong> a large number of serovars. The incubationperiod lasts from one <strong>to</strong> two weeks, although cases with an incubation periodof only two days or more than three weeks are known. The disease is characterizedby two phases, the bacteremic phase, lasting 7 <strong>to</strong> 10 days, <strong>and</strong> the lep<strong>to</strong>spiruricphase, lasting from a week <strong>to</strong> several months. Clinical manifestations vary <strong>and</strong> havediffering degrees of severity. In addition, numerous cases of infection occur inapparentlyor subclinically. In general, two clinical types are distinguished: icteric <strong>and</strong>anicteric. The serious icteric, or hepa<strong>to</strong>nephritic, type (Weil’s disease) is much lessfrequent than the anicteric type. Some authors estimate that this form occurs inapproximately 10% of cases. It is often associated with infection caused by icterohaemorrhagiae,but this is not the only serovar that can produce it. On the otherh<strong>and</strong>, numerous infections caused by icterohaemorrhagiae occur in anicteric form.In the classical form of Weil’s disease, the onset of symp<strong>to</strong>ms is sudden, with fever,headache, myalgias, conjunctivitis, nausea, vomiting, <strong>and</strong> diarrhea or constipation.Prostration may be severe. Petechiae on the skin, hemorrhages in the gastrointestinaltract, <strong>and</strong> proteinuria are <strong>common</strong>. Hepa<strong>to</strong>megaly <strong>and</strong> jaundice, renal insufficiencywith marked oliguria or anuria, azotemia, <strong>and</strong> electrolyte imbalance develop


LEPTOSPIROSIS 159with the disappearance of lep<strong>to</strong>spiremia <strong>and</strong> fever. If the patient’s conditionimproves, diuresis is reestablished <strong>and</strong> jaundice decreases. Convalescence lasts oneor two months, during which time fever, cephalalgia, myalgias, <strong>and</strong> general malaisemay reappear for a few days.In anicteric cases, the symp<strong>to</strong>ma<strong>to</strong>logy is milder. The symp<strong>to</strong>ms during lep<strong>to</strong>spiremia,which occurs during the first week of the disease, are fever, myalgias(particularly in the calves), conjunctivitis, stiffness in the neck, nausea, <strong>and</strong> sometimesvomiting. Often, the disease resembles influenza. The anicteric form has abenign course <strong>and</strong> patients recover in about a month. Lep<strong>to</strong>spiruria may continue fora week or several months after the disappearance of clinical symp<strong>to</strong>ms.Treatment should be started early in order <strong>to</strong> prevent tissue lesions. Penicillin G<strong>and</strong> amoxicillin were effective as late as one week after the onset of the disease(Benenson, 1990).The Disease in AnimalsCATTLE: At least 13 serovars have been isolated from cattle. In the Americas, thepredominant serovars in cattle are pomona, hardjo, <strong>and</strong> grippotyphosa; at times,infections caused by canicola <strong>and</strong> icterohaemorrhagiae, as well as by otherserovars, are found. The serovars pomona <strong>and</strong> hardjo are universal. As labora<strong>to</strong>rymethods have improved, outbreaks caused by the latter have been confirmed withincreasing frequency. In recent years, serovars belonging <strong>to</strong> the Hebdomadis grouphave been isolated more frequently. The importance of infection caused by someserovars is difficult <strong>to</strong> interpret. This is true of the serotype paidjan (Bataviaeserogroup), isolated from the kidneys of cattle (obtained in an Argentine slaughterhouse),<strong>and</strong> the serotype gal<strong>to</strong>ni (Canicola serogroup), isolated in Argentina <strong>and</strong>Colombia (Szyfres et al., 1967; Tedesco et al., 1969). To date, there are no knownoutbreaks caused by these serotypes in Argentina.The infection may cause an acute or subacute disease or remain clinically inapparent.The disease manifests with a fever lasting four or five days, anorexia, conjunctivitis,<strong>and</strong> diarrhea. Lep<strong>to</strong>spiremia begins <strong>to</strong> disappear when antibodies form, <strong>and</strong> thelep<strong>to</strong>spires completely disappear from the bloodstream in approximately one weekdue <strong>to</strong> humoral immunity. The surviving lep<strong>to</strong>spires are then harbored in the convolutedtubules of the kidneys <strong>and</strong> the infection enters a chronic phase. Lep<strong>to</strong>spiruriasheds enormous quantities of lep<strong>to</strong>spires <strong>to</strong> the outside environment, particularly duringthe first months of infection; later this decreases or ceases entirely. Lep<strong>to</strong>spiruriacaused by hardjo is much more prolonged than that caused by pomona. The hardjoserovar (Sejroe serogroup) in cattle is characterized by two syndromes: (a) agalactia,or a significant reduction in milk production, <strong>and</strong> (b) abortions or birthing of weakcalves that die soon after birth. In infections caused by hardjo—but not by pomona—it was found that lep<strong>to</strong>spires can reside in the genital tract (uterus <strong>and</strong> oviducts) in bothpregnant <strong>and</strong> nonpregnant females (Ellis <strong>and</strong> Thiermann, 1986). Infection of the genitaltract may indicate the possibility of sexual transmission (Prescott, 1991). L. interrogansis subdivided in<strong>to</strong> two genotypes: hardjo hardjo-bovis <strong>and</strong> hardjo hardjo-prajitno.The first genotype is the most prevalent in the US.Infertility may be a sequela of the infection. Serious cases include jaundice.However, the most notable symp<strong>to</strong>ms in a certain proportion of the animals are abortion<strong>and</strong> hemoglobinuria. Abortions usually occur between one <strong>and</strong> three weeksafter the onset of the disease. Up <strong>to</strong> 20% of aborting animals retain the placenta.


160 BACTERIOSESCattle of all ages are susceptible. The course of the disease is more severe incalves, which experience stunted growth <strong>and</strong> variable mortality rates.Quick-spreading epizootics are characterized by a high morbidity rate. It is possiblethat rapid passage of the lep<strong>to</strong>spires from one animal <strong>to</strong> another intensifiestheir virulence. In slow-moving epizootics, the rate of inapparent infection variesfrom one herd <strong>to</strong> another.Treatment with high doses of penicillin G or tetracycline is recommended foracute lep<strong>to</strong>spirosis. Dihydrostrep<strong>to</strong>mycin (12.5 mg/kg of bodyweight twice a day)may also be used, but due <strong>to</strong> its potential <strong>to</strong>xicity, treatment should be suspendedafter three days. Another suggested treatment is intramuscular sodium ampicillin (20mg/kg of bodyweight twice a day). In the chronic disease caused by pomona, it hasbeen repeatedly shown that a single intramuscular injection of dihydrostrep<strong>to</strong>mycin(25 mg/kg of bodyweight) eliminates the infection from the kidneys of most animalstreated. However, this treatment fails in the case of infection caused by hardjo,although the number of lep<strong>to</strong>spires is apparently reduced (Ellis et al., 1985).SWINE: The serovars most often isolated from swine in the Americas <strong>and</strong> in therest of the world are pomona, tarassovi, grippotyphosa, canicola, <strong>and</strong> icterohaemorrhagiae,as well as bratislava <strong>and</strong> muenchen of the Australis serogroup.Swine are a very important reservoir of pomona, with abundant <strong>and</strong> prolongedlep<strong>to</strong>spiruria. The clinical infection varies from one herd <strong>to</strong> another. In some cases,infection occurs subclinically, although the animals may exhibit a fever lasting a fewdays; in others, the infection produces such symp<strong>to</strong>ms as abortion <strong>and</strong> birth of weakpiglets. Stunted growth of piglets, jaundice, hemoglobinuria, convulsions, <strong>and</strong> gastrointestinaldisorders have also been seen. At times, meningitis <strong>and</strong> nervous symp<strong>to</strong>ma<strong>to</strong>logyare present. Abortion usually occurs between 15 <strong>and</strong> 30 days after infection.The principal serovars that cause abortions or stillborn piglets are pomona,tarassovi, <strong>and</strong> canicola. Infection that occurs during the last third of pregnancy isthe most critical in interrupting gestation. Lep<strong>to</strong>spires of the serovars bratislava <strong>and</strong>muenchen localize in the kidneys <strong>and</strong> in the genital tract of swine, as do hardjo lep<strong>to</strong>spiresin cattle.As in cattle, a single intramuscular injection of dihydrostrep<strong>to</strong>mycin (25 mg/kg ofbodyweight) is recommended for chronic infections caused by pomona.HORSES: Horses react serologically <strong>to</strong> many serotypes prevalent in the environment.Pomona has been isolated from these animals in the United States, <strong>and</strong> hardjohas been isolated in Argentina. In Europe, icterohaemorrhagiae, sejroe, <strong>and</strong> canicolahave been isolated, as well as pomona. Most infections are inapparent. Theremay be pho<strong>to</strong>phobia, watery eyes, edema of the ocular conjunctiva, miosis, <strong>and</strong> iritisin the acute phase of the disease. In the chronic phase, there may be anterior <strong>and</strong>posterior adhesions, a turbid vitreous body, formation of cataracts, uveitis, <strong>and</strong> otherophthalmologic abnormalities (Sillerud et al., 1987). Abortions may occasionallyoccur in infected mares (Bernard et al., 1993).Corneal opacity, which is frequently a sequela of the acute phase, can be reproducedthrough inoculation of inactivated lep<strong>to</strong>spires from various serovars. An antigenrelationship has also been demonstrated between L. interrogans, crystallin, <strong>and</strong>the cornea (Parma et al., 1986). Often, the disease’s sequela (periodic ophthalmia)is recognized instead of the acute, febrile phase. The onset of periodic ophthalmiaoccurs when the febrile phase has disappeared, after a latent phase that sometimes


LEPTOSPIROSIS 161lasts several months. Lep<strong>to</strong>spires have been detected in eye lesions of affected animals,<strong>and</strong> a high concentration of antibodies can be found in the aqueous humor.However, it should be borne in mind that lep<strong>to</strong>spirosis is not the only cause of periodicophthalmia. One hundred horses from the Minnesota River valley (USA) wereexamined ophthalmologically <strong>and</strong> serologically. A statistically significant associationwas found between uveitis <strong>and</strong> serology positive for pomona. Not all theseropositive horses were affected by uveitis, possibly due <strong>to</strong> different levels of exposure,strains of varying virulence, or different routes of infection (Sillerud et al.,1987). Serious cases of lep<strong>to</strong>spirosis with hepa<strong>to</strong>nephritic <strong>and</strong> cardiovascular syndromeshave been described in Europe.SHEEP AND GOATS: Epizootics in these species are not very frequent. Variousserovars that appear <strong>to</strong> have come from other animal species in the same environmenthave been isolated from sheep <strong>and</strong> goats in different countries (Faine, 1982),for example, hardjo in Australia <strong>and</strong> New Zeal<strong>and</strong>, pomona in the United States <strong>and</strong>New Zeal<strong>and</strong>, grippotyphosa in Israel, <strong>and</strong> ballum in Argentina. In WesternAustralia (Australia), a persistent lep<strong>to</strong>spirosis caused by the serovar hardjo wasfound in sheep that had no contact with cattle infected by the same serovar (Cousinset al., 1989). The authors conclude that, in addition <strong>to</strong> cattle, sheep could be a maintenancehost for hardjo.As in other ruminant species, the disease is characterized by fever, anorexia, <strong>and</strong>,in some animals, by jaundice, hemoglobinuria, anemia, abortion, birth of weak orstillborn animals, <strong>and</strong> infertility. The virulence of the infecting serovar <strong>and</strong> the conditionof the animal determine the severity of the clinical picture.DOGS AND CATS: The predominant serovars in dogs throughout the world are canicola<strong>and</strong> icterohaemorrhagiae. In addition <strong>to</strong> these serovars, pyrogenes, paidjan,<strong>and</strong> tarassovi have been isolated in Latin America <strong>and</strong> the Caribbean, <strong>and</strong> ballum,grippotyphosa, pomona, <strong>and</strong> bratislava have been isolated in the United States(Nielsen et al., 1991). Similar serovars predominate in Europe. The infection mayrange from asymp<strong>to</strong>matic <strong>to</strong> severe. The most serious form is the hemorrhagic,which begins suddenly with a fever that lasts from three <strong>to</strong> four days, followed bystiffness <strong>and</strong> myalgia in the hind legs, <strong>and</strong> hemorrhages in the oral cavity with a tendency<strong>to</strong>ward necrosis <strong>and</strong> pharyngitis. In a subsequent stage, there may be hemorrhagicgastroenteritis <strong>and</strong> acute nephritis. Jaundice may occur with infection bycanicola or by icterohaemorrhagiae, particularly in infection caused by the latterserovar. Case fatality is estimated at 10%.The disease rarely occurs in cats.WILD ANIMALS: Many wild animals, including rodents, are perfectly adapted <strong>to</strong>lep<strong>to</strong>spires <strong>and</strong> show no symp<strong>to</strong>ms or lesions.Source of Infection <strong>and</strong> Mode of Transmission (Figure 12): After a week oflep<strong>to</strong>spiremia, animals shed lep<strong>to</strong>spires in their urine, contaminating the environment.The best reservoirs of the infection are animals that have prolonged lep<strong>to</strong>spiruria<strong>and</strong> generally do not suffer from the disease themselves. For example, thisis true of rats, which harbor icterohaemorrhagiae <strong>and</strong> rarely have lesions. The infectionin man <strong>and</strong> animals is contracted directly or indirectly, through skin abrasions<strong>and</strong> the nasal, oral, <strong>and</strong> conjunctival mucosa. Indirect exposure through water, soil,or foods contaminated by urine from infected animals is the most <strong>common</strong> route. An


162 BACTERIOSESFigure 12. Lep<strong>to</strong>spirosis. Synanthropic transmission cycle.Infectedanimals(cattle, swine,rodents, dogs)Lep<strong>to</strong>spiruriaContamination ofsoil <strong>and</strong> water withurine containinglep<strong>to</strong>spiresSkin <strong>and</strong> mucusSusceptibleanimals(cattle, swine,rodents, dogs)Skin, oral <strong>and</strong>nasal mucosasManunusual case of transmission occurred in Great Britain, where an 11-year-old boyacquired the infection from a rat bite (Luzzi et al., 1987).People who work with lives<strong>to</strong>ck are often exposed <strong>to</strong> animal urine, directly or asan aerosol, which can contaminate the conjunctiva, nasal mucosa, or abrasions onexposed skin. They may also become infected indirectly by walking barefoot whereanimals have urinated. In many countries, domesticated animals, particularly swine<strong>and</strong> cattle, constitute important lep<strong>to</strong>spire reservoirs <strong>and</strong> a frequent source of infectionfor man.Rice-paddy workers are exposed <strong>to</strong> water contaminated by urine of rodents thatinfest the fields. Among agricultural workers, sugarcane harvesters are another highriskgroup. Field mice nesting among crops are a source of infection for harvesters,particularly during the early morning hours, when workers’ h<strong>and</strong>s come in<strong>to</strong> contactwith dew mixed with urine.Among pets, dogs are a <strong>common</strong> source of infection for man by serovars canicola<strong>and</strong> icterohaemorrhagiae.Tropical regions are endemic areas of lep<strong>to</strong>spirosis <strong>and</strong> the highest case rates correspond<strong>to</strong> areas with heavy rainfall. The highest number of cases occurs during therainy season. Epidemic outbreaks erupt because of environmental changes, such asflooding, which cause rodents <strong>to</strong> move in<strong>to</strong> cities. An example of this is the epidemicsthat occurred in the city of Recife (Pernambuco State, Brazil), in 1966 <strong>and</strong>1970, with 181 <strong>and</strong> 102 cases, respectively. The predominant serovar was icterohaemorrhagiae.Humidity, high temperatures, <strong>and</strong> an abundance of rats were theprincipal fac<strong>to</strong>rs that precipitated these outbreaks as well as others in tropicalregions. Small epidemic outbreaks are also caused by recreational activities, such asswimming or diving in streams or ponds contaminated by the urine of infected animals.An outbreak occurred in a cattle- <strong>and</strong> swine-raising region of Cuba, where 21cases were diagnosed in people who bathed in the Clavellina River <strong>and</strong> theManiadero reservoir. The Pomona <strong>and</strong> Australis serogroups were predominant <strong>and</strong>two isolates of the latter were obtained from the river water (Suárez Hernández et


LEPTOSPIROSIS 163al., 1989). Epidemic outbreaks caused by several different serovars have occurredamong soldiers wading in streams or camping by riverbanks during jungle maneuvers.Such epidemics have occurred in Panama <strong>and</strong> Malaysia; in these cases, thesource of infection was the urine of infected wild animals.Animals, either primary or secondary hosts, contract the infection in a similarway. The density of the host population <strong>and</strong> the environmental conditions in whichit lives play important roles. On cattle ranches, the infection is usually introduced bya carrier animal with lep<strong>to</strong>spiruria <strong>and</strong>, at times, by fields that flood with water contaminatedat a neighboring establishment.Pathogenic lep<strong>to</strong>spires (L. interrogans) do not multiply outside the animal organism.Consequently, in addition <strong>to</strong> carrier animals, existence of a lep<strong>to</strong>spirosis focusrequires environmental conditions favorable <strong>to</strong> the survival of the agent in the exteriorenvironment. Lep<strong>to</strong>spires need high humidity, a neutral or slightly alkaline pH,<strong>and</strong> suitable temperatures. Low, inundated ground <strong>and</strong> artificial or natural freshwaterreceptacles (ponds, streams, reservoirs, etc.) are favorable <strong>to</strong> their survival,whereas salt water is deleterious. Soil composition, both its physiochemical <strong>and</strong> biologicalcharacteristics (microbe population), also acts <strong>to</strong> prolong or shorten life forlep<strong>to</strong>spires in the environment. Temperatures in the tropics constitute a very favorablefac<strong>to</strong>r for survival of lep<strong>to</strong>spires, but cases of lep<strong>to</strong>spirosis may also occur incold climates, although they are less frequent.Role of Animals in the Epidemiology of the Disease: Wild <strong>and</strong> domesticatedanimals are essential for the maintenance of pathogenic lep<strong>to</strong>spires in nature.Transmission of the infection from animals <strong>to</strong> man is effected directly or indirectly.Human-<strong>to</strong>-human transmission is rare. Man is an accidental host <strong>and</strong> only in veryspecial conditions can he contribute <strong>to</strong> the maintenance of an epidemic outbreak.Such was the case in an epidemic described in the forest northeast of Hanoi(Vietnam). The outbreak occurred among soldiers occupied in logging <strong>and</strong> transpor<strong>to</strong>f logs by buffalo through a swampy area. Lep<strong>to</strong>spiruria was observed in 12% of 66convalescent soldiers. In contrast, the rate of infection was insignificant among buffalo<strong>and</strong> rodents in the region. The pH of the surface water was neutral, the workersworked barefoot, <strong>and</strong> their urine had a pH that fluctuated around seven (their dietwas vegetarian). Lep<strong>to</strong>spiruria persisted in some of the soldiers for more than sixmonths (Spinu et al., 1963).A case of transmission through mother’s milk was described in the US (Songer<strong>and</strong> Thiermann, 1988). A female veterinarian continued nursing after being infectedwith the serovar hardjo while performing an au<strong>to</strong>psy on a cow. Twenty-one daysafter the appearance of clinical symp<strong>to</strong>ms in the mother, the baby became ill withfever, anorexia, irritability, <strong>and</strong> lethargy. The serovar hardjo was isolated from thebaby’s urine <strong>and</strong> the baby recovered with antibiotic treatment.Various cases of congenital infection have also been described (Faine, 1991).Diagnosis: In man, the etiologic agent can be isolated from blood during the firstweek of the disease; afterwards, it can be isolated from the urine, either by directculture or by inoculation in<strong>to</strong> young hamsters. Repeated blood samples are necessaryfor serological examination. The patient has no antibodies during the first week;they appear in six <strong>to</strong> seven days <strong>and</strong> reach maximum levels in the third or fourthweek. If the first sample is negative or low-titer <strong>and</strong> the second shows an appreciableincrease in antibody titer (fourfold or more), lep<strong>to</strong>spirosis is indicated.


164 BACTERIOSESThe same diagnostic procedures are employed for animals as for man. Blood orurine may be used for the bacteriologic examination, depending on the stage of theillness. If a necropsy is performed on a sacrificed or dead animal, kidney culturesshould be made. Examination of several tissue samples from the same individual isnot always easily done in veterinary practice, but individual diagnosis of domesticanimals is not as important as herd diagnosis. Discovery of high antibody titers inseveral members of a herd <strong>and</strong> a clinical picture compatible with lep<strong>to</strong>spirosis indicatea recent infection.Low titers may indicate residual antibodies from a past infection or recentlyformed antibodies that have not yet had time <strong>to</strong> reach a high level.The serologic reference test that is used most for man as well as for animals ismicroscopic agglutination (MAT). This test should be carried out using representativeserovars from different serogroups, especially those occurring in the region. Itis necessary <strong>to</strong> bear in mind that cross-reactions are produced not only betweendifferent serovars of the same serogroup but, at the beginning of the infection(two <strong>to</strong> three weeks), also between serovars of different serogroups, <strong>and</strong> a heterologousserovar titer may predominate. Reaction <strong>to</strong> the homologous serovarbecomes more pronounced with time. Cross-reactions are much more frequent inman than in animals.The macroscopic plate test with inactivated antigens can be used as a preliminaryor screening test for man <strong>and</strong> animals. It is fast <strong>and</strong> easy, <strong>and</strong> particularly useful fordiagnosing disease in a herd.Plate agglutination is a genus-specific test, which uses as an antigen the pa<strong>to</strong>cstrain of saprophytic lep<strong>to</strong>spira (L. biflexa) <strong>to</strong> determine if the patient is sufferingfrom lep<strong>to</strong>spirosis (Mazzonelli et al., 1974). Reaction <strong>to</strong> this test is marked duringthe acute phase of lep<strong>to</strong>spirosis <strong>and</strong> then quickly becomes negative (Faine, 1982).Among more recent tests, those of interest are indirect immunofluorescence<strong>and</strong> enzyme-linked immunosorbent assay (ELISA). With both, the types ofimmunoglobulins (IgM or IgG) can be determined by using the correspondingantigens. IgM appears after the first week of the disease <strong>and</strong> IgG appears afterseveral weeks. In some human cases, IgG antibodies cannot be detected for reasonsas yet unknown.The utility of ELISA for diagnosing infection due <strong>to</strong> hardjo was compared withMAT. It was found that a positive reaction can be obtained with MAT 10 days afterthe animal has been experimentally infected; ELISA does not give a positive reactionuntil 25 days after infection. In addition, there was a 90% concordance betweenboth tests. Cross-reactions with sera from animals inoculated with other serotypesoccurred in fewer than 1% (Bercovich et al., 1990).The serovar hardjo is subdivided in<strong>to</strong> subserovars or genotypes: hardjo genotypehardjo-bovis <strong>and</strong> hardjo genotype prajitno. A DNA probe for genotype hardjo-boviswas developed by LeFebvre (1987). Three methods for detecting hardjo type hardjoboviswere compared: with DNA hybridization, 60 of the 75 urine samples fromcows experimentally exposed were positive; with immunofluorescence, 24 sampleswere positive; <strong>and</strong> with culturing, only 13 were positive. The DNA probe was shown<strong>to</strong> be much more sensitive than the other techniques in detecting the genotypehardjo-bovis (Bolin et al., 1989a).A very sensitive generic test is polymerase chain reaction (PCR), which can detectas few as 10 lep<strong>to</strong>spires (Mérien et al., 1992).


LEPTOSPIROSIS 165Control: In man, control measures include: (a) personal hygiene; (b) use of protectiveclothes during farm work; (c) drainage of lowl<strong>and</strong>s whenever possible; (d)rodent-proof structures; (e) food protection <strong>and</strong> correct garbage disposal; (f) controlof infection in domestic animals; (g) avoidance of swimming in streams <strong>and</strong> otherfresh watercourses that may be contaminated, <strong>and</strong> (h) chemoprophylaxis of highriskoccupational groups (sugarcane harvesters, rice-paddy workers, or soldiers).Human immunization has not been widely applied. It has been used with promisingresults in Italy, Pol<strong>and</strong>, <strong>and</strong> the former Soviet Union. However, because of secondary,mainly allergic effects, its use did not spread. Tests of a vaccine made in achemically defined, protein-free medium are under way (Shenberg <strong>and</strong> Torten,1973). In China, a similar vaccine is being used on a wide scale.The use of antibiotics in prophylaxis <strong>and</strong> treatment of human lep<strong>to</strong>spirosis hasyielded contradic<strong>to</strong>ry results. One study (Takafuji et al., 1984) showed that doxycyclineis effective in chemoprophylaxis; the same drug is probably also effective intreatment. Because lep<strong>to</strong>spirosis caused many cases of disease among American soldierstraining in Panama, a double-blind field test was undertaken <strong>to</strong> determine theefficacy of doxycycline in preventing the infection. Nine hundred forty soldier volunteerswere r<strong>and</strong>omly divided in<strong>to</strong> two groups. One group was given an oral doseof 200 mg of doxycycline each week for three weeks, <strong>and</strong> the other group was givena placebo. After remaining in the jungle for three weeks, 20 cases of lep<strong>to</strong>spirosiswere diagnosed in the placebo group (attack rate of 4.2%) <strong>and</strong> only one case wasdiagnosed in the doxycycline group (attack rate of 0.2%), i.e., the drug was 95%effective (Takafuji et al., 1984). It has been suggested (Sanford, 1984) that chemoprophylaxiswould be justified in areas where incidence is 5% or higher.Mechanization of farm work has resulted in a decrease of outbreaks, for example,among rice-paddy workers.Among domesticated animals, vaccination of pigs, cattle, <strong>and</strong> dogs is effective inpreventing the disease, but it does not protect completely against infection.Vaccinated animals may become infected without showing clinical symp<strong>to</strong>ms; theymay have lep<strong>to</strong>spiruria, although <strong>to</strong> a lesser degree <strong>and</strong> for a shorter time thanunvaccinated animals. A few known human cases of lep<strong>to</strong>spirosis were contractedfrom vaccinated dogs. There are bacterins <strong>to</strong> protect against the pomona, hardjo, <strong>and</strong>grippotyphosa serovars in cattle; against pomona in swine; <strong>and</strong> against canicola <strong>and</strong>icterohaemorrhagiae in dogs. Immunity is predominantly serovar-specific, <strong>and</strong> theserovar or serovars active in a focus must be known in order <strong>to</strong> correctly immunizethe animals. Females should be vaccinated before the reproductive period <strong>to</strong> protectthem during pregnancy. Young animals can be immunized after 3 or 4 months of age.Bacterins now in use require annual revaccination. For herds <strong>to</strong> which outside animalsare being introduced, it is recommended that vaccination be repeated every sixmonths. An effective measure is <strong>to</strong> combine vaccination with antibiotic treatment(Thiermann, 1984).Vaccination against hardjo is not very satisfac<strong>to</strong>ry, not even if the prevalent genotypehardjo-bovis is used in combined vaccines (Bolin et al., 1989b) or in monovalentvaccines (Bolin et al., 1991).It has been demonstrated that vaccination with bacterins initially stimulates theproduction of IgM antibodies, which disappear after a few months <strong>and</strong> are replacedby IgG antibodies. Vaccination generally does not interfere with diagnosis becauseof the quick disappearance of IgM antibodies, which are active in agglutination. IgG


166 BACTERIOSESare the protective antibodies <strong>and</strong> can be detected with serum protection assays inhamsters or with the growth inhibition test in culture media.A vaccine derived from the outer membrane of lep<strong>to</strong>spires has been obtained <strong>and</strong>has yielded very promising results in labora<strong>to</strong>ry tests by conferring resistance no<strong>to</strong>nly against the disease but also against the establishment of lep<strong>to</strong>spiruria.Chemotherapy is promising. Experiments have shown that a single injection of dihydrostrep<strong>to</strong>mycinat a dose of 25 mg/kg of bodyweight is effective against lep<strong>to</strong>spiruriain cattle <strong>and</strong> swine. The infection has been eradicated in several herds withantibiotic treatment <strong>and</strong> proper environmental hygiene. The combination of vaccination<strong>and</strong> chemotherapy for the control of swine lep<strong>to</strong>spirosis has been proposed.Proper herd management is important for control. It has been repeatedly demonstratedthat swine can transmit the pomona serovar <strong>to</strong> cattle. Therefore, separationof these two species is important for prophylaxis.BibliographyAlex<strong>and</strong>er, A.D. Lep<strong>to</strong>spira. In: Balows, A., W.J. Hausler, K.L. Hermann, H.D. Isenberg,H.J. Shadomy, eds. Manual of Clinical Microbiology. 5th ed. Washing<strong>to</strong>n, D.C.: AmericanSociety for Microbiology; 1991.Alex<strong>and</strong>er, A.D., W.E. Gochenour, Jr., K.R. Reinhard, M.K. Ward, R.H. Yagen.Lep<strong>to</strong>spirosis. In: Bodily, H.L., ed. Diagnostic Procedures for Bacterial, Mycotic <strong>and</strong>Parasitic Infections. 5th ed. New York: American Public Health Association, 1970.Als<strong>to</strong>n, J.M., J.C. Broom. Lep<strong>to</strong>spirosis in Man <strong>and</strong> Animals. Edinburgh, London:Livings<strong>to</strong>ne; 1958.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bercovich, Z., R. Taaijke, B.A. Bokhout. Evaluation of an ELISA for the diagnosis ofexperimentally induced <strong>and</strong> naturally occurring Lep<strong>to</strong>spira hardjo infections in cattle. VetMicrobiol 21:255–262, 1990.Bernard, W.V., C. Bolin, T. Riddle, et al. Lep<strong>to</strong>spiral abortion <strong>and</strong> lep<strong>to</strong>spiruria in horsesfrom the same farm. J Am Vet Med Assoc 202:1285–1286, 1993.Bolin, C.A., J.A. Cassells, R.L. Zuerner, G. Trueba. Effect of vaccination with a monovalentLep<strong>to</strong>spira interrogans serovar hardjo type hardjo-bovis vaccine on type hardjo-bovisinfection of cattle. Am J Vet Res 52:1639–1643, 1991.Bolin, C.A., R.L. Zuerner, G. Trueba. Comparison of three techniques <strong>to</strong> detect Lep<strong>to</strong>spirainterrogans serovar hardjo type hardjo-bovis in bovine urine. Am J Vet Res 50:1001–1003, 1989a.Bolin, C.A., R.L. Zuerner, G. Trueba. Effect of vaccination with a pentavalent lep<strong>to</strong>spiralvaccine containing Lep<strong>to</strong>spira interrogans serovar hardjo type hardjo-bovis on type hardjobovisinfection in cattle. Am J Vet Res 50:2004–2008, 1989b.Cacchione, R.A. Enfoques de los estudios de la lep<strong>to</strong>spirosis humana y animal en AméricaLatina. Rev Asoc Argent Microbiol 5:36–53, 100–111, 143–154, 1973.Cousins, D.V., T.M. Ellis, J. Parkinson, C.H. McGlashan. Evidence for sheep as a maintenancehost for Lep<strong>to</strong>spira interrogans serovar hardjo. Vet Rec 124:123–124, 1989.Diesch, S.L., H.C. Ellinghausen. Lep<strong>to</strong>spiroses. In: Hubbert, W.T., W.F. McCulloch, P.R.Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield:Thomas; 1975.Ellis, W.A., J. Montgomery, J.A. Cassells. Dihydrostrep<strong>to</strong>mycin treatment of bovine carriersof Lep<strong>to</strong>spira interrogans serovar hardjo. Res Vet Sci 39:292–295, 1985.


LEPTOSPIROSIS 167Ellis, W.A., A.B. Thiermann. Isolation of lep<strong>to</strong>spires from the genital tracts of Iowa cows.Am J Vet Res 47:1694–1696, 1986.Everard, C.O., A.E. Green, J.W. Glosser. Lep<strong>to</strong>spirosis in Trinidad <strong>and</strong> Grenada, with specialreference <strong>to</strong> the mongoose. Trans Roy Soc Trop Med Hyg 70:57–61, 1976.Faine, S., ed. Guidelines for the control of lep<strong>to</strong>spirosis. Geneva: World HealthOrganization; 1982. (Offset Publication 67).Faine, S. Lep<strong>to</strong>spirosis. In: Evans, A.S., P.S. Brachman, eds. Bacterial Infections ofHumans. 2nd ed. New York: Plenum Medical Book Co.; 1991.Hanson, L.E., D.N. Tripathy, A.H. Killinger. Current status of lep<strong>to</strong>spirosis immunizationin swine <strong>and</strong> cattle. J Am Vet Med Assoc 161:1235–1243, 1972.Hart, R.J., J. Gallagher, S. Waitkins. An outbreak of lep<strong>to</strong>spirosis in cattle <strong>and</strong> man. BritMed J (Clin Res Ed) 288(6435):1983–1984, 1984.LeFebvre, R.B. DNA probe for detection of the Lep<strong>to</strong>spira interrogans serovar hardjogenotype hardjo-bovis. J Clin Microbiol 25:2236–2238, 1987.Lep<strong>to</strong>spirosis in man, British Isles, 1983. Br Med J (Clin Res Ed) 288(6435):1984–1985, 1984.Luzzi, G.A., L.M. Milne, S.A. Waitkins. Rat-bite acquired lep<strong>to</strong>spirosis. J Infect 15:57–60, 1987.Mazzonelli, J. Advances in bovine lep<strong>to</strong>spirosis. Bull Off Int Epizoot 3:775–808, 1984.Mazzonelli, J., G.T. Dorta de Mazzonelli, M. Mailloux. Possibilité de diagnostiquesérologique des lep<strong>to</strong>spires á l’aide d’un antigène unique. Med Mal Infect 4:253, 1974.Mérien, F., P. Amouriaux, P. Perolat, et al. Polymerase chain reaction for detection ofLep<strong>to</strong>spira spp. in clinical samples. J Clin Microbiol 30:2219–2224, 1992.Myers, D.M. Serological studies <strong>and</strong> isolations of serotype hardjo <strong>and</strong> Lep<strong>to</strong>spira biflexastrains from horses of Argentina. J Clin Microbiol 3:548–555, 1976.Nielsen, J.N., G.K. Cochran, J.A. Cassells, L.E. Hanson. Lep<strong>to</strong>spira interrogans serovarbratislava infection in two dogs. J Am Vet Med Assoc 199:351–352, 1991.Parma, A.E., C.G. Santisteban, A.S. Fernández, et al. Relación antigénica entre Lep<strong>to</strong>spirainterrogans, cristalino y córnea equina, probada por enzimoinmunoensayo. Rev Med Vet67:72–76, 1986.Perolat, P., F. Grimont, F. Regnault, et al. rRNA gene restriction patterns of Lep<strong>to</strong>spira:Amolecular typing system. Res Microbiol 141:159–171, 1990.Prescott, J. Treatment of lep<strong>to</strong>spirosis [edi<strong>to</strong>rial]. Cornell Vet 81:7–12, 1991.Sanford, J.P. Lep<strong>to</strong>spirosis—time for a booster. N Engl J Med 310:524–525, 1984.Shenberg, E., M. Torten. A new lep<strong>to</strong>spiral vaccine for use in man. I. Development of a vaccinefrom Lep<strong>to</strong>spira grown on a chemically defined medium. J Infect Dis 128:642–646, 1973.Sillerud, C.L., R.F. Bey, M. Ball, S.I. Bistner. Serologic correlation of suspected Lep<strong>to</strong>spirainterrogans serovar pomona-induced uveitis in a group of horses. J Am Vet Med Assoc191:1576–1578, 1987.Songer, J.G., A.B. Thiermann. Lep<strong>to</strong>spirosis. J Am Vet Med Assoc 193:1250–1254, 1988.Spinu, I., V. Topcin, Trinh Thi Hang Quy, Vo Van Hung, Mguyen Sy Quoe, Chu Xnan Long,et al. L’homme comme réservoir de virus dans une épidémie de lep<strong>to</strong>spirose survenue dans lajungle. Arch Roum Path Exp 22:1081–1100, 1963.Stalheim, O.H. Vaccination against lep<strong>to</strong>spirosis: Protection of hamsters <strong>and</strong> swine againstrenal lep<strong>to</strong>spirosis by killed but intact gamma-irradiated or dihydrostrep<strong>to</strong>mycin-exposedLep<strong>to</strong>spira pomona. Am J Vet Res 28:1671–1676, 1967.Stalheim, O.H. Chemotherapy of renal lep<strong>to</strong>spirosis in cattle. Am J Vet Res 30:1317–1323, 1969.Stalheim, O.H. Duration of immunity in cattle in response <strong>to</strong> a viable, avirulent Lep<strong>to</strong>spirapomona vaccine. Am J Vet Res 32:851–854, 1971.Suárez Hernández, M., J. Bustelo Aguila, V. Gorgoy González, et al. Estudio epidemiológicode un brote de lep<strong>to</strong>spirosis en bañistas en el poblado de Jicotea de la provincia Ciegode Ávila. Rev Cubana Hig Epidemiol 27:272–284, 1989.


168 BACTERIOSESSulzer, C.R., W.L. Jones. Lep<strong>to</strong>spirosis Methods in Labora<strong>to</strong>ry Diagnosis. Atlanta: U.S.Centers for Disease Control <strong>and</strong> Prevention; 1974.Szyfres, B. La lep<strong>to</strong>spirosis como problema de salud humana y animal en América Latinay el área del Caribe. In: VIII Reunión Interamericana sobre el Control de la Fiebre Af<strong>to</strong>sa yOtras Zoonosis. Washing<strong>to</strong>n, D.C.: Organización Panamericana de la Salud; 1976.(Publicación Científica 316).Szyfres, B., C.R. Sulzer, M.M. Gal<strong>to</strong>n. A new lep<strong>to</strong>spiral serotype in the Bataviaeserogroup from Argentina. Trop Geogr Med 19:344–346, 1967.Takafuji, E.T., J.W. Kirkpatrick, R.N. Miller, et al. An efficacy trial of doxycycline chemoprophylaxisagainst lep<strong>to</strong>spirosis. N Engl J Med 310:497–500, 1984.Tedesco, L.F., G. Manrique, C.R. Sulzer. A new lep<strong>to</strong>spiral serotype in the Canicolaserogroup from Argentina. Trop Geogr Med 21:203–206, 1969.Thiermann, A.B. Lep<strong>to</strong>spirosis: Current developments <strong>and</strong> trends. J Am Vet Med Assoc184:722–725, 1984.Tripathy, D.N., A.R. Smith, L.E. Hanson. Immunoglobulins in cattle vaccinated with lep<strong>to</strong>spiralbacterins. Am J Vet Res 36:1735–1736, 1975.Van der Hoeden, J. Lep<strong>to</strong>spirosis. In: Van der Hoeden, J., ed. Zoonoses. Amsterdam:Elsevier; 1964.Waitkins, S.A. From the PHLS. Update on lep<strong>to</strong>spirosis. Brit Med J (Clin Res Ed)290(6480):1502–1503, 1985.World Health Organization. Research needs in lep<strong>to</strong>spirosis [memor<strong>and</strong>um]. Bull WorldHealth Organ 47:113–122, 1972.LISTERIOSISICD-10 A32.1 listerial meningitis <strong>and</strong> meningoencephalitis;A32.7 listerial septicaemia; A32.8 other forms of listeriosis;P37.2 neonatal (disseminated) listeriosisSynonyms: Leukocy<strong>to</strong>sis, listerial infection, listeriasis, listerellosis, circling disease(in animals).Etiology: The genus Listeria contains seven species, but only two are of interestin human <strong>and</strong> animal pathology: L. monocy<strong>to</strong>genes <strong>and</strong> L. ivanovii (formerly L. bulgaricaor serovar 5 of L. monocy<strong>to</strong>genes). A notable difference between the twopathogenic species is their hemolytic ability.The most important species for both man <strong>and</strong> animals is L. monocy<strong>to</strong>genes, agram-positive, facultatively anaerobic bacillus 0.5 <strong>to</strong> 2 microns long <strong>and</strong> 0.5microns in diameter that is motile at temperatures between 20°C <strong>and</strong> 25°C. It isbeta-hemolytic in blood agar <strong>and</strong> forms a narrow b<strong>and</strong> of hemolysis around thecolonies (unlike L. ivanovii, which forms a wide b<strong>and</strong>). A noteworthy characteristicof L. monocy<strong>to</strong>genes is its ability <strong>to</strong> grow at low temperatures; at a pH between 6<strong>and</strong> 9, it can reproduce at temperatures from 3°C <strong>to</strong> 45°C. It is a facultative, intracellularparasite of the reticuloendothelial system. For purposes of epidemiological


LISTERIOSIS 169research, L. monocy<strong>to</strong>genes is subdivided in<strong>to</strong> 11 serovars. Most human (92%) <strong>and</strong>animal cases are caused by serovars 4b, 1/2b, <strong>and</strong> 1/2a (Bor<strong>to</strong>lussi et al., 1985).Therefore, serotyping is of limited usefulness for identifying a source of infection(Gelin <strong>and</strong> Broome, 1989).Of 161 isolates serotyped in the US, 33% belonged <strong>to</strong> serovar 4b; 31.5%, <strong>to</strong>serovar 1/2b; 30%, <strong>to</strong> serovar 1/2a; 4%, <strong>to</strong> serovar 3b; 1%, <strong>to</strong> serovar 3a; <strong>and</strong> 0.5%,<strong>to</strong> serovar 1/2c (Gelin et al., 1987). When 71 isolates were serotyped in Brazil, sevendifferent serovars were recognized; 50% were 4b <strong>and</strong> 29.6% were 1/2a (Hofer et al.,1984).Although serotyping has been useful as a preliminary approach, other schemeshad <strong>to</strong> be devised in order <strong>to</strong> be able <strong>to</strong> specify the source of infection. Subtypingwas done primarily with two methods: phage typing <strong>and</strong> electrophoretic enzymetyping. In Great Britain, 64% of the strains could be typed using 28 phages; inFrance, 78% could be typed using 20 phages. In both cases, a sizeable percentage ofstrains could not be typed. All strains of L. monocy<strong>to</strong>genes can be typed using theisoenzymatic subtyping method (Sel<strong>and</strong>er et al., 1986). Recently, ribosomal RNAtyping (ribotyping) has been used with success.Geographic Distribution: Worldwide. L. monocy<strong>to</strong>genes is widely distributed invegetation, soil, <strong>and</strong> human <strong>and</strong> animal intestines.Occurrence in Man: Incidence is low, but it is an important disease because ofits high mortality. In many developing countries, listeriosis is rare. There is greaterconcentration of cases in European countries <strong>and</strong> the United States, perhaps becausemedical personnel in these countries are more on the lookout for the disease <strong>and</strong>because better labora<strong>to</strong>ry support is available. In the former Federal Republic ofGermany (West Germany), there were 296 cases of listeriosis between 1969 <strong>and</strong>1985, 60% of which occurred in urban areas. Fifty percent of the strains isolatedwere from newborns <strong>and</strong> the most <strong>common</strong> serovar was 4b (Schmidt-Wolf et al.,1987). In Great Britain, information was obtained from 722 cases occurring from1967 <strong>to</strong> 1985. In 246 cases (34%), the infection affected the mother, the fetus, or thenewborn. Neonatal infection was diagnosed within the first two days postpartum in133 (54%) cases; 56 cases (23%) were diagnosed later than two days postpartum.There were 47 (19%) cases of intrauterine death. There were also 10 cases (4%) ofmothers with bacteremia that did not affect the fetus. Overall case fatality was 50%(McLauchlin, 1990a). The author estimates that these data must include less than50% of the <strong>to</strong>tal number of cases that occurred in the country. In adults <strong>and</strong> youth,474 cases were recorded; 275 (58%) were men <strong>and</strong> 199 (42%) were nonpregnantwomen. Case fatality was 44%. Seventy-six percent of these patients had an underlyingillness. An increase in incidence was noted in autumn (McLauchlin, 1990b).There were an estimated 800 cases per year in the US between 1980 <strong>and</strong> 1982,with an incidence of 3.6 per million inhabitants <strong>and</strong> at least 150 deaths (19% casefatality). The highest attack rates were seen in newborns (4.7 per 100,000 live births)<strong>and</strong> in persons 70 years of age or older (11 per million) (Gellin <strong>and</strong> Broome, 1989).There are few recognized cases in developing countries. Sporadic cases have beenseen in several Latin American countries. In a Mexican hospital, hemocultures werecarried out during a three-month period on all children whose mothers showed signsof amniotic infection; L. monocy<strong>to</strong>genes was isolated from 4 out of 33 newbornsexamined (Pérez-Mirabate <strong>and</strong> Giono, 1963). In Peru (Guevara et al., 1979),


170 BACTERIOSESserovars 4d <strong>and</strong> 4b of L. monocy<strong>to</strong>genes were isolated from three fatal cases ofneonatal listeriosis <strong>and</strong> from five aborted fetuses. In Argentina, there are few data onthe occurrence of human listeriosis. In Córdoba (Argentina), there are cases ofneonatal listeriosis each year, <strong>and</strong> these constitute between 2% <strong>and</strong> 3% of bacteriologicallyconfirmed sepsis (Paolasso, 1981). In a small <strong>to</strong>wn in the province ofBuenos Aires, Manzullo (1981) isolated L. monocy<strong>to</strong>genes type 1a from a bovinefetus, the vaginal exudate of the woman who milked the cows, <strong>and</strong> from the household’sfemale dog. In another <strong>to</strong>wn, Manzullo (1990) isolated the agent from awoman’s vaginal exudate <strong>and</strong> from the woman’s female cat. In a Buenos Aires medicalcenter, nine cases of listeriosis were diagnosed in 15 years, two of them fatal.Only one patient was not immunocompromised (Roncoroni et al., 1987).Most cases occur sporadically, but epidemic outbreaks have occurred in severalcountries. In 1981, in a maternity hospital in Halifax (Canada), there were 34 perinatalcases <strong>and</strong> 7 cases in women without underlying illness or immunosuppression.Case fatality in the babies born live was 27%. There were five spontaneous abortions<strong>and</strong> four babies stillborn at term. The epidemic outbreak was attributed <strong>to</strong> coleslaw:L. monocy<strong>to</strong>genes serovar 4b was isolated from the cabbage as well as from thepatients. On the farm where the cabbages were grown, two sheep had died from listeriosisthe previous year; in addition, the farmer used sheep dung as fertilizer. It isalso worth noting that the farmer kept the cabbage refrigerated at 4°C, whichallowed the etiologic agent <strong>to</strong> multiply at the expense of other contaminant microorganisms(Schlech et al., 1983).An earlier outbreak occurred in 1979 in eight hospitals in Bos<strong>to</strong>n (USA); itaffected 20 patients, 15 of whom acquired the infection in the hospital. Raw vegetableswere assumed <strong>to</strong> be the source of infection.In Massachusetts (USA), an epidemic outbreak caused by pasteurized milk wasrecorded in 1983. It affected 42 immunocompromised patients <strong>and</strong> seven immunocompetentpatients; there were also perinatal cases. Case fatality was 29%. Of 40isolates, 32 were type 4b. It is possible that the milk had been contaminated afterpasteurization (Schuchat et al., 1991).The largest epidemic in the US was recorded in 1985 in Los Angeles, California(Linnan et al., 1988). The epidemic affected pregnant women, their fetuses, <strong>and</strong>their newborns. Case fatality was 63% for the infected fetuses <strong>and</strong> newborns. Theoutbreak was due <strong>to</strong> a Mexican soft cheese; serovar 4b was isolated from the patients<strong>and</strong> the cheese. The incubation period was 11 <strong>to</strong> 70 days, with an average of 31 days(Schuchat et al., 1991).There have been epidemic outbreaks in Switzerl<strong>and</strong>, Denmark, <strong>and</strong> France. InSwitzerl<strong>and</strong>, the 1987 outbreak that led <strong>to</strong> 64 perinatal cases <strong>and</strong> 58 nonperinatalcases was caused by a soft cheese. Case fatality was 28%. The strain of L. monocy<strong>to</strong>genesresponsible was the same enzymatic type as the strain that caused the outbreakin California in 1985 (Gelin <strong>and</strong> Broome, 1989). One of the largest epidemicsknown <strong>to</strong> date occurred in France in 1992. It affected 691 persons <strong>and</strong> 40% of thecases were caused by serotype 4b. The epidemic strain was isolated from 91 pregnantwomen <strong>and</strong> their children. Of the remaining persons affected by the epidemicstrain, 61% were immunodeficient. The phage type was the same as in California(1985), Switzerl<strong>and</strong> (1983–1987), <strong>and</strong> Denmark (1985–1987). The epidemic strainwas isolated from 163 samples of meat products, 35 cheese samples, <strong>and</strong> 12 otherfood samples. The epidemic lasted from 18 March <strong>to</strong> 23 December 1992 <strong>and</strong> caused


LISTERIOSIS 17163 deaths <strong>and</strong> 22 abortions. The cause was attributed <strong>to</strong> pig’s <strong>to</strong>ngue in gelatin(<strong>WHO</strong>, 1993). In 1993 (January–August), 25 new cases occurred in France. Thistime the outbreak was again due <strong>to</strong> serogroup 4, but <strong>to</strong> a different lysotype than inthe 1992 epidemic. Of the 25 cases, 21 were maternal-fetal, with 4 spontaneousabortions <strong>and</strong> 2 stillbirths at term. Most of the cases occurred in western France. Theepidemiological investigation was able <strong>to</strong> attribute the infection <strong>to</strong> a pork product(“rillettes”) distributed by a single commercial firm (Bol Epidemiol Hebdom No.34, 1993).There are various risk groups: pregnant women, fetuses, newborns, the elderly,<strong>and</strong> immunocompromised patients. However, there was some controversy regardingAIDS patients. Several authors maintained that AIDS patients were unlikely <strong>to</strong> contractlisteriosis, even though their cellular immunity system was highly compromised.While it is true that listeriosis is not one of the principal conditions affectingAIDS patients, its incidence in those infected by HIV is 300 times higher than in thegeneral population (Schuchat et al., 1991).Occurrence in Animals: Listeriosis has a wide variety of domestic <strong>and</strong> wild animalhosts. The infection has been confirmed in a large number of domestic <strong>and</strong> wildmammals, in birds, <strong>and</strong> even in poikilotherms. The most susceptible domesticspecies is sheep, followed by goats <strong>and</strong> cattle. The frequency of occurrence in theseanimals is not known.Outbreaks in sheep have been described in several Latin American countries. Thedisease has been confirmed in alpacas in Peru, <strong>and</strong> in sheep, fowl, <strong>and</strong> cattle inArgentina <strong>and</strong> Uruguay.The first epizootic outbreak (1924) was recognized in Engl<strong>and</strong> in labora<strong>to</strong>ry rabbitssuffering from a disease characterized by mononucleosis, from whence the specificname of the agent, monocy<strong>to</strong>genes, comes. Mononucleosis rarely occurs inman or in animals other than rabbits <strong>and</strong> rodents.Several outbreaks have been described in Great Britain <strong>and</strong> the US due <strong>to</strong> silagewith a pH higher than 5, which favors multiplication of L. monocy<strong>to</strong>genes. As theuse of silage increases, outbreaks, which occur when the quality of silage is poor <strong>and</strong>the pH high, increase as well.The Disease in Man: The most affected group is newborns (50% of cases inFrance <strong>and</strong> 39% in the US), followed by those over age 50. The disease is very rarebetween 1 month <strong>and</strong> 18 years of age. According <strong>to</strong> data from two German obstetricalclinics, listerial infection caused 0.15% <strong>to</strong> 2% of perinatal mortality. Listerialabortion in women usually occurs in the second half of pregnancy, <strong>and</strong> is more frequentin the third trimester. Symp<strong>to</strong>ms that precede miscarriage or birth by a fewdays or weeks may include chills, increased body temperature, cephalalgia, slightdizziness, <strong>and</strong> sometimes, gastrointestinal symp<strong>to</strong>ms. These septicemic episodesmay or may not recur before birth of a stillborn fetus or a seriously ill full-term baby.After delivery, the mother shows no disease symp<strong>to</strong>ms, but L. monocy<strong>to</strong>genes canbe isolated from the vagina, cervix, <strong>and</strong> urine for periods varying from a few days<strong>to</strong> several weeks. If the child is born alive but was infected in utero, it may showsymp<strong>to</strong>ms immediately after birth or within a few days. The symp<strong>to</strong>ma<strong>to</strong>logy is tha<strong>to</strong>f sepsis or, less frequently, a disseminated granuloma<strong>to</strong>sis (granuloma<strong>to</strong>sis infantisepticum).There may also be symp<strong>to</strong>ms of a respira<strong>to</strong>ry tract disorder. Case fatalityis high. The main lesion is a focal hepatic necrosis in the form of small, grayish-


172 BACTERIOSESwhite nodules. Some children born apparently healthy fall ill with meningitis shortlythereafter (a few days <strong>to</strong> several weeks). In these cases, the infection was probablyacquired in utero or during birth. In the US, neonatal meningitis is the most <strong>common</strong>clinical form, while in Europe, perinatal septicemia prevails. Hydrocephalus isa <strong>common</strong> sequela of neonatal meningitis.Meningitis or meningoencephalitis is the most <strong>common</strong> clinical form in adults,especially in those over 50. Listerial meningitis often occurs as a complication indebilitated persons, alcoholics, diabetics, in patients with neoplasias, or in elderlypatients with a declining immune system. Before the existence of antibiotics, casefatality was 70%. Listerial septicemia also occurs among weakened adults, especiallypatients undergoing long-term treatment with corticosteroids or antimetabolites.In addition, listeriosis may result in endocarditis, external <strong>and</strong> internalabscesses, <strong>and</strong> endophthalmitis. A cutaneous eruption has been described amongveterinarians who h<strong>and</strong>led infected fetuses.The recommended treatment for maternal-fetal listeriosis is ampicillin. Variousantibiotics, such as ampicillin (alone or in combination with aminoglycosides),tetracycline (not for those under 8 years of age), <strong>and</strong> chloramphenicol, may be usedfor the other forms of the disease (Benenson, 1990).The Disease in AnimalsSHEEP, GOATS, AND CATTLE: Listeriosis manifests itself in ruminants as encephalitis,neonatal mortality, <strong>and</strong> septicemia. The most <strong>common</strong> clinical form is encephalitis. Insheep <strong>and</strong> goats, the disease has a hyperacute course, <strong>and</strong> mortality may vary from 3%<strong>to</strong> more than 30%. In cattle, listerial encephalitis has a chronic course, with the animalssurviving for 4 <strong>to</strong> 14 days. In general, only 8% <strong>to</strong> 10% of a herd is affected.A ruminant with encephalitis isolates itself from the herd <strong>and</strong> shows symp<strong>to</strong>ms ofdepression, fever, lack of coordination, <strong>to</strong>rticollis, spasmodic contractions <strong>and</strong> paralysisof facial muscles <strong>and</strong> throat, profuse salivation, strabismus, <strong>and</strong> conjunctivitis.The animal tries <strong>to</strong> lean against some support while st<strong>and</strong>ing <strong>and</strong>, if able <strong>to</strong> walk,moves in circles. In the final phase of the disease, the animal lies down <strong>and</strong> makescharacteristic chewing movements when attempting <strong>to</strong> eat.Listerial encephalitis can affect animals of any age, but it is more <strong>common</strong> in thefirst three years of life. Nevertheless, it does not appear before the rumen becomesfunctional. Septicemia is much more <strong>common</strong> in young animals than adults.Abortion occurs mainly during the last months of gestation <strong>and</strong> is generally the onlysymp<strong>to</strong>m of genital infection, the dam showing no other signs of disease. If uterineinfection occurs in the cow before the seventh month of pregnancy, the dead fetus isusually retained in the uterus for several days <strong>and</strong> has a macerated appearance, withmarked focal necrotic hepatitis. In addition, the placenta may be retained <strong>and</strong> metritusmay develop. If infection occurs in the final months of pregnancy, the fetus ispractically intact <strong>and</strong> shows minimal lesions.L. monocy<strong>to</strong>genes can also cause mastitis in cows. There are few described cases,either because the presence of this agent in cows has not been studied or because itsoccurrence really is rare. Mastitis caused by Listeria varies in severity from subclinical<strong>to</strong> acute <strong>and</strong> chronic. Elimination of the agent in milk occurs over a longperiod of time <strong>and</strong> may have public health repercussions, especially since pasteurizationdoes not guarantee complete safety if the viable bacteria count is high beforeheat treatment (Gitter, 1980).


LISTERIOSIS 173A study carried out in 1970–1971 in Vic<strong>to</strong>ria (Australia) (Dennis, 1975) showedthat listeriosis is an important cause of perinatal mortality in sheep. In 94 flocks,fetuses <strong>and</strong> lambs that died during the neonatal period were examined, <strong>and</strong> L. monocy<strong>to</strong>geneswas found in 25%. The disease caused by this agent occurs mostly in winter.It has been estimated that the rate of abortion in flocks affected by listeriosis inVic<strong>to</strong>ria varies from 2% <strong>to</strong> 20%.L. ivanovii, which differs from L. monocy<strong>to</strong>genes on the basis of several phenotypiccharacteristics, was associated in several countries with abortions in sheep <strong>and</strong>,occasionally, in cows (Alex<strong>and</strong>er et al., 1992).OTHER MAMMALS: Listeriosis is rare in swine; when it does occur in the first fewweeks of life, it usually takes the septicemic form. Few cases are known in dogs, inwhich the disease may be confused with rabies. In other domestic <strong>and</strong> wild species,the disease generally appears as isolated cases <strong>and</strong> in the septicemic form.Outbreaks have been described in rabbit <strong>and</strong> guinea pig breeding colonies.FOWL: Young birds are the most affected. Outbreaks are infrequent <strong>and</strong> mortalitymay range from the loss of a few birds on one farm <strong>to</strong> a high rate of losses onother farms. The septicemic form is the most <strong>common</strong>, with degenerative lesions ofthe myocardium, pericarditis, <strong>and</strong> focal hepatic necrosis. On rare occasions, themeningoencephalitic form is found, with marked <strong>to</strong>rticollis. Since the generalizeduse of antibiotics in poultry feed began, few cases of listeriosis in this species havebeen reported.Source of Infection <strong>and</strong> Mode of Transmission: The causal agent is widely distributedin animals <strong>and</strong> man, as well as in the environment. L. monocy<strong>to</strong>genes hasbeen isolated from different mammalian <strong>and</strong> avian species <strong>and</strong> from the soil, plants,mud, pasture, wastewater, <strong>and</strong> streams. The presence of virulent <strong>and</strong> avirulent (formice) strains in animals <strong>and</strong> in the environment complicates clarification of the epidemiology,but serotyping can be of considerable help. Cattle, sheep, <strong>and</strong> manyother animal species eliminate the agent in their feces. L. monocy<strong>to</strong>genes has alsobeen isolated from the feces of patients <strong>and</strong> their contacts, as well as from a smallpercentage of the general human population. However, it has been isolated from thes<strong>to</strong>ols of some 20% <strong>to</strong> 30% of pregnant women, <strong>and</strong> has also been found in thefemale genital tract. In addition <strong>to</strong> untypeable strains, potentially pathogenicserotype 1 <strong>and</strong> serovar 4b have been isolated (Kampelmacher <strong>and</strong> van NoorleJansen, 1980). Consequently, the natural reservoir is wide <strong>and</strong> the number of hostsis large. Despite this, few people contract the disease. Many women from whoses<strong>to</strong>ols the agent has been isolated give birth <strong>to</strong> healthy children. Concurrent conditions,such as stress <strong>and</strong> other predisposing causes (particularly <strong>diseases</strong> or treatmentsthat depress the immune system), come in<strong>to</strong> play in initiating the disease.Another predisposing cause is the decline in the immune system that occurs withaging, as well as endocrine changes during pregnancy <strong>and</strong> deficiencies inimmunoregulation at the placental level.The source of infection for the fetus <strong>and</strong> newborn is evidently the infected motherherself. It is believed that the almost inapparent disease course manifested by themother is caused by a mild bacteremia. Airborne infection might play a role, as suggestedby the influenza-like symp<strong>to</strong>ms exhibited by the mother. The mother’s genitaltract is probably infected via the fecal route, while the fetus is infected via the


174 BACTERIOSESbloodstream or placenta. The discovery of the causal agent in the semen of a manwhose wife’s genitals were infected would indicate that, in some cases, the infectionmay be transmitted through sexual contact.The oral route of transmission seems <strong>to</strong> be important, as indicated by the recen<strong>to</strong>utbreaks occurring in the US, Switzerl<strong>and</strong>, <strong>and</strong> France (see the section on occurrencein man), where some contaminated vegetables, milk <strong>and</strong> milk products, <strong>and</strong>meat were the vehicle of the infection. It is also interesting <strong>to</strong> note that the milk thatled <strong>to</strong> one of the outbreaks came from establishments where listeriosis had beendiagnosed in the animals. The disease affected two very susceptible groups: newborns<strong>and</strong> debilitated persons. Of 49 patients hospitalized with listerial septicemiaor meningitis, 7 were newborns <strong>and</strong> 42 were adults. All the adults were sufferingfrom other <strong>diseases</strong> or undergoing treatment with immunosuppressants.A rise in listeriosis cases when animals feed on silage would indicate the digestivesystem as the portal of entry. The causal agent has been isolated from poorlyprepared fodder that had a pH higher than 5. During an outbreak of encephalitis insheep, the same serovar <strong>and</strong> phage type of L. monocy<strong>to</strong>genes was isolated from thesilage <strong>and</strong> from the animals’ brains. The silage contained 1 million listeriae per gram(Vázquez-Bol<strong>and</strong> et al., 1992).L. monocy<strong>to</strong>genes is distributed in populations of healthy animals <strong>and</strong> the diseasecan be produced when stress lowers the host’s resistance.Although it has been demonstrated that food has been the source of infection inboth human <strong>and</strong> animal outbreaks, the source of infection is not known with certaintyin sporadic cases in man. However, it has been possible <strong>to</strong> confirm that a significantpercentage of such cases were caused by the ingestion of a contaminatedfood (Schuchat et al., 1992; Pinner et al., 1992). Cases of listeriosis have been associatedwith ingestion of raw sausage <strong>and</strong> undercooked chicken (Schwartz et al.,1988). It is likely that many cases with a food source cannot be detected because theextended incubation period makes it impossible for patients <strong>to</strong> associate a food withtheir infection (Gelin <strong>and</strong> Broome, 1989).One case of listeriosis in a woman with cancer was associated with consumptionof turkey franks. The investigation established that the franks in opened packages inher refrigera<strong>to</strong>r were contaminated, but those in unopened packages were not.Cultures from other foods in the refrigera<strong>to</strong>r also yielded positive results. The conclusionwas that a cross-contamination was involved (CDC, 1989).An interesting study was conducted in the US in 1992 (CDC, 1992). During theperiod 1988–1990, special epidemiological surveillance was conducted in four of thecountry’s districts, with a population of 18 million inhabitants. There were 301 casesof listeriosis identified (7.4 per million inhabitants), 67 (23%) of whom died. Thepatients’ food consumption his<strong>to</strong>ries indicated that the listeriosis patients ingested 2.6times more soft cheeses than did the controls, or purchased 1.6 times more preparedfoods. The patients’refrigera<strong>to</strong>rs were examined <strong>and</strong> it was found that 79 (64%) of 123contained at least one food contaminated by L. monocy<strong>to</strong>genes; in 26 (33%) of the 79refrigera<strong>to</strong>rs the same enzymatic strain as that found in the patients was isolated.The wide distribution of Listeria spp. in nature <strong>and</strong> in animal feces explains whyits presence in raw meats is almost inevitable. Prevalence in raw meats may varyfrom 0% <strong>to</strong> 68%. Pork is contaminated most often, but contamination is also frequentin uncooked chicken. There is little information regarding the virulence of L.monocy<strong>to</strong>genes strains isolated from meats (Johnson et al., 1990).


LISTERIOSIS 175There is no uniform criterion regarding when <strong>to</strong> reject foods according <strong>to</strong> thedegree of contamination by L. monocy<strong>to</strong>genes. Several countries (France, US)require that there be no contamination at all, while others (Canada, Germany) havea certain <strong>to</strong>lerance. It is impossible <strong>to</strong> ensure the <strong>to</strong>tal absence of Listeria spp. in allfoods (Dehaumont, 1992).In California (USA), a six-month study was conducted on the prevalence ofListeria spp. in environmental samples from 156 milk-processing plants. Listeriaspp. was isolated from 75 (12.6%) of the 597 environmental samples. Half of theisolates were identified as L. monocy<strong>to</strong>genes. Of the 156 plants, 46 gave positivesamples for Listeria spp. <strong>and</strong> 19.9% of these isolates were identified as L. monocy<strong>to</strong>genes(Charl<strong>to</strong>n et al., 1990).Role of Animals in the Epidemiology of the Disease: The epidemiology of sporadiclisteriosis is still not well known. Most researchers consider it a disease <strong>common</strong><strong>to</strong> man <strong>and</strong> animals <strong>and</strong> not a zoonosis per se. It is likely that animals contribute<strong>to</strong> maintenance of listeria in general in nature <strong>and</strong> especially <strong>to</strong> its distribution.Studies conducted in recent years suggest that man <strong>and</strong> animals can contract theinfection from many sources. Most cases in man occur in urban areas, where thereis little contact with animals. Nonetheless, animals may be the source of the infection.In one case, infection was confirmed in a woman who drank raw milk; the sameserotype of Listeria was isolated from the raw milk <strong>and</strong> from the woman’s prematuretwins. The etiologic agent was isolated from 16% of cows that had listerialabortions. The previously described outbreaks caused by milk, meat, or vegetablescontaminated by manure from listeria-infected animals demonstrate that animalsmay be an important source of infection.There are indisputable cases of direct transmission of the infection from animals<strong>to</strong> man. A cattleman assisted during the delivery of a cow, inserting his arms in theuterus. Within the next 24 hours, a rash appeared on his h<strong>and</strong>s <strong>and</strong> one arm <strong>and</strong>developed in<strong>to</strong> pustules. He later experienced fever, chills, <strong>and</strong> generalized pain. Thesame phage type was isolated from the cow’s vagina <strong>and</strong> from the cattleman’s pustules(Cain <strong>and</strong> McCann, 1986). The veterinary profession is particularly at risk ofcontracting cutaneous listeriosis. Many veterinarians have become ill after attendingcows that aborted, fetuses, or newborns, or after conducting au<strong>to</strong>psies of septicemicanimals. The most frequent lesion is a papular exanthema (Owen et al., 1960;Nieman <strong>and</strong> Lorber, 1980; Hird, 1987). Contact with sick birds may also causehuman infection (Gray <strong>and</strong> Killinger, 1966).Diagnosis: Diagnosis can be made only through isolation of the causal agent. Ifthe sample is obtained from usually sterile sites, such as blood, cerebrospinal fluid,amniotic fluid, or biopsy material, seeding can be done directly in blood agar, withincubation at 35°C for a week <strong>and</strong> daily checks. Listeria can be isolated from anyorgan in septicemic fetuses.In sheep, goats, or cattle with encephalitis, samples of the medulla oblongatashould be cultured. In septicemic fowl, rodents, or neonatal ruminants, blood orinternal organs should be cultured. The “cold enrichment” method is used especiallyin epidemiological investigations <strong>and</strong> is indicated for culturing highly contaminatedspecimens. However, this method has no diagnostic value for clinical cases becauseof the time it takes, since treatment with antibiotics (preferably ampicillin) shouldbegin as soon as possible <strong>to</strong> be effective.


176 BACTERIOSESAt present, contaminated samples as well as foods are cultured in an enrichmentmedium <strong>and</strong> then in a selective medium. One procedure is the US Department ofAgriculture procedure; it uses nalidixic acid <strong>and</strong> acriflavin in broth <strong>to</strong> inhibit thegrowth of contaminating flora. The culture is incubated for 24 hours at 30°C <strong>and</strong>then a subculture is done in another broth of the same composition for another 24hours at 30°C. Finally, a highly selective solid medium that contains lithium chloride<strong>and</strong> moxalactam is used (McClain <strong>and</strong> Lee, 1988).A test has been developed <strong>to</strong> distinguish pathogenic from nonpathogenic strainsof L. monocy<strong>to</strong>genes. This method is based on the potentiating <strong>and</strong> synergistic effectthat the extrosubstance of Rhodococcus equi has for producing hemolysis in culturesof pathogenic strains of L. monocy<strong>to</strong>genes (Skalka et al., 1982).In general, serologic tests are confusing <strong>and</strong> not useful because of cross-reactionswith enterococci <strong>and</strong> Staphylococcus aureus, especially by serogroups 1 <strong>and</strong> 3 ofListeria. DNA probes that specifically detect L. monocy<strong>to</strong>genes have been developed(Datta et al., 1988).Control: In regions where human neonatal listeriosis is <strong>common</strong>, a Gram staincan be made from the meconium of a newborn, <strong>and</strong> treatment with antibiotics canbe rapidly initiated if bacteria suspected of being Listeria are found. Women whodevelop influenza-like symp<strong>to</strong>ms in the final months of pregnancy should be carefullyexamined <strong>and</strong> treated, if necessary, with antibiotics. The limited arsenal ofdefense against the infection includes such measures as the pasteurization of milk,rodent control, <strong>and</strong> <strong>common</strong> practices of environmental <strong>and</strong> personal hygiene.Special recommendations have been developed for food preparation (CDC,1992): cook products of animal origin well, thoroughly wash vegetables thatare eaten raw, keep raw meats separate from other foods, do not consume rawmilk, wash utensils used in food preparation well, <strong>and</strong> reheat all food lef<strong>to</strong>vers at ahigh temperature. Immunocompromised individuals must not eat soft cheeses <strong>and</strong>veterinarians must take precautions during delivery, <strong>and</strong> particularly during abortions<strong>and</strong> au<strong>to</strong>psies.Animals with encephalitis or those that have aborted should be isolated <strong>and</strong> theirplacentas <strong>and</strong> fetuses destroyed. Recently acquired animals should only be added <strong>to</strong>a herd after undergoing a reasonable period of quarantine.BibliographyAlex<strong>and</strong>er, A.V., R.L. Walker, B.J. Johnson, et al. Bovine abortions attributable <strong>to</strong> Listeriaivanovii: Four cases (1988–1990). J Am Vet Med Assoc 200:711–714, 1992.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bojsen-Moller, J. Human listeriosis: Diagnostic, epidemiological <strong>and</strong> clinical studies. ActaPathol Microbiol Sc<strong>and</strong> (Suppl 229):1–157, 1972.Bor<strong>to</strong>lussi, R., W.F. Schlech III, W.L. Albrit<strong>to</strong>n. Listeria. In: Lennette, E.H., A. Balows,W.J. Hausler, Jr., H.J. Shadomy. Manual of Clinical Microbiology. 4th ed. Washing<strong>to</strong>n, D.C.:American Society for Microbiology; 1985.Broadbent, D.W. Infections associated with ovine perinatal mortality in Vic<strong>to</strong>ria. Aust Vet J51:71–74, 1975.


LISTERIOSIS 177Cain, D.B., V.L. McCann. An unusual case of cutaneous listeriosis. J Clin Microbiol23:976–977, 1986.Charl<strong>to</strong>n, B.R., H. Kinde, L.H. Jensen. Environmental survey for Listeria species inCalifornia milk processing plants. J Food Protect 53:198–201, 1990.Datta, A.R., B.A. Wentz, D. Shook, M.W. Trucksess. Synthetic oligodeoxyribonucleotideprobes for detection of Listeria monocy<strong>to</strong>genes. Appl Environ Microbiol 54:2933–2937, 1988.Dehaumont, P. Listeria monocy<strong>to</strong>genes et produits alimentaires: “zéro<strong>to</strong>lérance au moins.”Bull Epidemiol Hebdom 24:109, 1992.Dennis, S.M. Perinatal lamb mortality in Western Australia. 6. Listeric infection. Aust Vet J51:75–79, 1975.Fleming, D.W., S.L. Cochi, K.L. MacDonald, J. Brondum, P.S. Hayes, B.D. Plikaytis, et al.Pasteurized milk as a vehicle of infection in an outbreak of listeriosis. N Engl J Med312(7):404-407, 1985.Franck, M. Contribution a l’Étude de l’Épidémiologie des Listerioses Humaines etAnimales [thesis]. École Nationale Vétérinaire de Lyon, France, 1974.García, H., M.E. Pin<strong>to</strong>, L. Ross, G. Saavedra. Brote epidémico de listeriosis neonatal. RevChil Pediatr 54:330–335, 1983.Gellin, B.G., C.V. Broome. Listeriosis. JAMA 261:1313–1320, 1989.Gellin, B.G., C.V. Broome, A.W. High<strong>to</strong>wer. Geographic differences in listeriosis in theUnited States [abstract]. 27th Interscience Conference of Antimicrobial Agents <strong>and</strong>Chemotherapy. New York, Oct 5, 1987. Cited in: Gellin, B.G., C.V. Broome. Listeriosis.JAMA 261:1313–1320, 1989.Gitter, M., R. Bradley, P.H. Blampied. Listeria monocy<strong>to</strong>genes infection in bovine mastitis.Vet Rec 107:390–393, 1980.Gray, M.L., A.H. Killinger. Listeria monocy<strong>to</strong>genes <strong>and</strong> listeric infections. Bact Rev30:309–382, 1966.Green, H.T., M.B. Macaulay. Hospital outbreak of Listeria monocy<strong>to</strong>genes septicaemia: Aproblem of cross infection? Lancet 2:1039–1040, 1978.Guevara, J.M., J. Pereda, S. Roel. Human listeriosis in Peru. Tropenmed Parasi<strong>to</strong>l30:59–61, 1979.Hird, D.W. Review of evidence for zoonotic listeriosis. J Food Protect 50:429–433, 1987.Hofer, E., G.V.A. Pessoa, C.E.A. Melles. Listeriose humana. Prevalência dos sorotipos deListeria monocy<strong>to</strong>genes isolados no Brasil. Rev Inst Adolfo Lutz 44:125–131, 1984.Johnson, J.L., M.P. Doyle, R.G. Cassens. Listeria monocy<strong>to</strong>genes <strong>and</strong> other Listeria spp. inmeat <strong>and</strong> meat products. A review. J Food Protect 53:81–91, 1990.Kampelmacher, E.H., L.M. van Noorle Jansen. Listeriosis in humans <strong>and</strong> animals in theNetherl<strong>and</strong>s (1958–1977). Zbl Bakt Hyg Orig A 246:211–227, 1980.Killinger, A.H. Listeriosis. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger, eds.Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.Killinger, A.H., M.E. Mansfield. Epizootiology of listeric infection in sheep. J Am Vet MedAssoc 157:1318–1324, 1970.Larsen, H.E. Epidemiology of listeriosis. The ubiqui<strong>to</strong>us occurrence of Listeria monocy<strong>to</strong>genes.In: Proceedings, Third International Symposium on Listeriosis, Bilthoven, TheNetherl<strong>and</strong>s, 1966.Linnan, M.J., L. Mascola, X.D. Lou, et al. Epidemic listeriosis associated with Mexicanstylecheese. N Engl J Med 319:823–828, 1988.Mair, N.S. Human listeriosis. In: Graham-Jones, O., ed. Some Diseases of AnimalsCommunicable <strong>to</strong> Man in Britain. Oxford: Pergamon; 1968.Manzullo, A. Epidemiología y epizootiología de la listeriosis. Acta Bioq Clin Latinoam14:539–546, 1981.Manzullo, A.C. Listeriosis [letter]. An Acad Nacl Agr Vet 44 (4):5–13, 1990.McClain, D., W.H. Lee. Development of USDA-FSIS method for isolation of Listeriamonocy<strong>to</strong>genes from raw meat <strong>and</strong> poultry. J Assoc Off Anal Chem 71:660–664, 1988.


178 BACTERIOSESMcLauchlin, J. Human listeriosis in Britain, 1967–85, a summary of 722 cases. 1.Listeriosis during pregnancy <strong>and</strong> in the newborn. Epidemiol Infect 104:181–190, 1990a.McLauchlin, J. Human listeriosis in Britain, 1967–85, a summary of 722 cases. 2.Listeriosis in non-pregnant individuals, a changing pattern of infection <strong>and</strong> seasonal incidence.Epidemiol Infect 104:191–201, 1990b.Moro, M. Enfermedades infecciosas de las alpacas. 2. Listeriosis. Rev Fac Med Vet16/17:154–159, 1961–1962.Nieman, R.E., B. Lorber. Listeriosis in adults: A changing pattern. Report of eight cases <strong>and</strong>review of the literature, 1968–1978. Rev Infect Dis 2:207–227, 1980.Owen, C.R., A. Neis, J.W. Jackson, H.G. S<strong>to</strong>enner. A case of primary cutaneous listeriosis.N Engl J Med 362:1026–1028, 1960.Paolasso, M.R. Listeria en Córdoba. Acta Bioq Clin Latinoam 14:581–584, 1981.Pérez-Miravete, A., S. Giono. La infección perinatal listérica en México. II. Aislamien<strong>to</strong> deListeria monocy<strong>to</strong>genes en septicemia del recién nacido. Rev Inst Salubr Enferm Trop23:103–113, 1963.Pinner, R.W., A. Schuchat, B. Swaminathan, et al. Role of foods in sporadic listeriosis. II.Microbiologic <strong>and</strong> epidemiologic investigation. The Listeria Study Group. JAMA267:2046–2050, 1992.Rocourt, J., J.M. Alonso, H.P. Seeliger. Virulence comparée des cinq groupes génomiquesde Listeria monocy<strong>to</strong>genes (sensu la<strong>to</strong>). Ann Microbiol 134A:359–364, 1983.Roncoroni, A.J., M. Michans, H.M. Bianchini, et al. Infecciones por Listeria monocy<strong>to</strong>genes.Experiencia de 15 años. Medicina 47:239–242, 1987.Schlech, W.F., P.M. Lavigne, R.A. Bor<strong>to</strong>lussi, et al. Epidemic listeriosis—evidence fortransmission by food. N Engl J Med 308: 203–206, 1983.Schmidt-Wolf, G., H.P.R. Seeliger, A. Schretten-Brunner. Menschliche Listeriose-Erkrankungen in der Bundesrepublik Deutschl<strong>and</strong>, 1969–1985. Zbl Bakt Hyg A 265:472–486, 1987.Schuchat, A., K.A. Deaver, J.D. Wenger, et al. Role of foods in sporadic listeriosis. I. Casecontrolstudy of dietary risk fac<strong>to</strong>rs. JAMA 267:2041–2045, 1992.Schuchat, A., B. Swaminathan, C.V. Broome. Epidemiology of human listeriosis. ClinMicrobiol Rev 4:169–183, 1991.Schwartz, B., C.A. Ciesielski, C.V. Broome, et al. Association of sporadic listeriosis withconsumption of uncooked hot dogs <strong>and</strong> undercooked chicken. Lancet 2:779–782, 1988.Seeliger, H.P.R. Listeriosis. New York: Hafner Publishing Co.; 1961.Sel<strong>and</strong>er, R.K., D.A. Caugant, H. Ochman, et al. Methods of multilocus enzyme electrophoresisfor bacterial population genetics <strong>and</strong> systematics. Appl Environ Microbiol51:873–884, 1986.Skalka, B., J. Smola, K. Elischerova. Routine test for in vitro differentiation of pathogenic<strong>and</strong> apathogenic Listeria monocy<strong>to</strong>genes strains. J Clin Microbiol 15:503–507, 1982.Stamm, A.M., W.E. Dismukes, B.P. Simmons, C.G. Cobbs, A. Elliott, P. Budrich, et al.Listeriosis in renal transplant recipients: Report of an outbreak <strong>and</strong> review of 102 cases. RevInfect Dis 4:665–682, 1982.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Listeriosis associated with consumption of turkey franks.MMWR Morb Mort Wkly Rep 38(15):267–268, 1989.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Update: Foodborne listeriosis—United States, 1988–1990.MMWR Morb Mort Wkly Rep 41:251, 257–258, 1992.Vázquez-Bol<strong>and</strong>, J.A., L. Domínguez, M. Blanco, et al. Epidemiologic investigation of asilage-associated epizootic of ovine listeric encephalitis, using a new Listeria-selective enumerationmedium <strong>and</strong> phage typing. Am J Vet Res 53:368–371, 1992.Weis, J., H.P.R. Seeliger. Incidence of Listeria monocy<strong>to</strong>genes in nature. Appl Microbiol30:29–32, 1975.


LYME DISEASE 179World Health Organization (<strong>WHO</strong>). Outbreak of listeriosis in 1992. Wkly Epidemiol Rec68(13):89–91, 1993.Young, S. Listeriosis in cattle <strong>and</strong> sheep. In: Faulkner, L.C., ed. Abortion Diseases ofLives<strong>to</strong>ck. Springfield: Thomas; 1968.LYME DISEASEICD-10 A69.2Synonyms: Lyme borreliosis, Lyme arthritis, erythema migrans (formerly erythemachronicum migrans) with polyarthritis.Etiology: The etiologic agent is a spirochete, transmitted by ticks of the Ixodesricinus complex <strong>and</strong> named Borrelia burgdorferi in honor of the person who discoveredit (Burgdorfer et al., 1982; Steere et al., 1983; Johnson et al., 1984). Thegenus Borrelia belongs <strong>to</strong> the family Spirochaetaceae <strong>and</strong> is made up of spiralshaped,actively motile bacteria. B. burgdorferi is 11 <strong>to</strong> 39 microns long <strong>and</strong> has 7<strong>to</strong> 11 flagella. The strains of B. burgdorferi isolated in Europe have demonstratedsome heterogeneity, particularly in the two principal plasmid-dependent surface proteins(Steere, 1990).Geographical Distribution <strong>and</strong> Occurrence in Man: The human disease hasbeen recognized in 46 states in the US. Areas with endemic foci in that country arethe Atlantic coast (particularly in the Northeast), Wisconsin <strong>and</strong> Minnesota in theMidwest, <strong>and</strong> California <strong>and</strong> Oregon along the Pacific coast (Benenson, 1990). Thenatural foci of the infection are exp<strong>and</strong>ing. In New York State, the number of countieswith recorded human cases increased from four <strong>to</strong> eight between 1985 <strong>and</strong> 1989<strong>and</strong> the number of counties where the presence of the tick Ixodes dammini, the vec<strong>to</strong>rof the infection, was documented increased from 4 <strong>to</strong> 22 during the same period(White et al., 1991). 1 In the US, more than 40,000 cases were recorded between1982 <strong>and</strong> 1992, <strong>and</strong> it is currently the principal disease transmitted by ticks. Themajor vec<strong>to</strong>rs of the infection in the US are Ixodes dammini in the East <strong>and</strong>Midwest, <strong>and</strong> I. pacificus on the Pacific coast.The etiological agent has also been isolated in Ontario (Canada). Many Europeancountries record cases of Lyme borreliosis <strong>and</strong> the vec<strong>to</strong>r on that continent is Ixodesricinus. The disease has also been recognized in Australia, China, Japan, <strong>and</strong> coun-1A study indicates that Ixodes dammini <strong>and</strong> I. scapularis (a tick in the southern US) aregeographic variants of the same species, which would correctly be named I. scapularis (Oliveret al., 1993). Since there are differences in terms of ecology <strong>and</strong> the rate of infection(Kazmierczak <strong>and</strong> Sorhage, 1993), we feel it is advisable <strong>to</strong> retain the terminology <strong>common</strong>lyin use for both varieties in order <strong>to</strong> avoid confusion.


180 BACTERIOSEStries of the former Soviet Union. In the Asian countries, the tick that transmits theinfection is I. persulcatus.In the Northern Hemisphere, the disease has the highest incidence in summer duringthe months of June <strong>and</strong> July, but it may appear in other seasons depending onthe tick life cycle in the region (Benenson, 1990).Occurrence in Animals: In endemic areas <strong>and</strong> areas near <strong>to</strong> them, various speciesof domestic animals (dogs, horses, <strong>and</strong> cattle) are infected by B. burgdorferi.In the natural foci of the infection, wild animals form the major part of the lifecycle of the tick <strong>and</strong> of the agent it transmits. In these foci, high rates of reac<strong>to</strong>rs <strong>to</strong>the indirect immunofluorescence test, using antigens from the etiologic agent, havebeen found in several wild animal species. The prevalence of reac<strong>to</strong>rs among animalsinfested with I. dammini in eastern Connecticut from 1978 <strong>to</strong> 1982 was as follows(Magnarelli et al., 1984): white-tailed deer (Odocoileus virginianus), 27%;white-footed mice (Peromyscus leucopus), 10%; eastern chipmunks (Tamias striatus),17%; gray squirrels (Sciurus carolinensis), 50%; opossums (Didelphis virginiana),17%; raccoons (Procyon lo<strong>to</strong>r), 23%; <strong>and</strong> dogs, 24%. The spirochete was isolatedfrom the bloodstream of 1 out of 20 white-footed mice examined (Anderson<strong>and</strong> Magnarelli, 1983; Bosler et al., 1984).Of 380 samples obtained from dogs from two locations selling animals inWisconsin, 53% reacted positively <strong>to</strong> the immunofluorescence test <strong>and</strong> the pathogenicagent was isolated from the blood of 8 out of 111 dogs (Burgess, 1986). In Texas, thesame test was used <strong>to</strong> examine 2,409 canine samples in 1988; of these, 132 (5.5%)yielded positive results. Many of the seropositive dogs were from the north-centralpart of the state, where most of the human cases are recorded (Cohen et al., 1990).It has been noted that horses are frequently bitten by I. dammini. In a serologicalstudy of 50 r<strong>and</strong>omly selected horses in New Engl<strong>and</strong> (USA), a known endemicarea, 13 of the horses were reactive <strong>to</strong> the indirect immunofluorescence test.In another serological survey using the enzyme-linked immunosorbent assay(ELISA) technique, 13 of 100 horses examined in the month of June tested positive<strong>and</strong> 6 of 91 (7%) tested positive in the month of Oc<strong>to</strong>ber. The horses came from fiveeastern US states. The frequency of antibody responses was higher in horses fromNew Jersey than in horses from Pennsylvania (Bernard et al., 1990). In contrast, noreactive horses were found in central Texas (Cohen et al., 1992).The Disease in Man: The characteristic cutaneous lesion, erythema migrans(EM), appears from 3 <strong>to</strong> 20 days after the tick bite. The lesion begins with a red maculaor papule that widens. The borders are clearly delineated, the central lesionpales, <strong>and</strong> an annular erythema forms. The erythema may be recurrent, with secondarylesions appearing on other parts of the body. The cutaneous lesions may beaccompanied for several weeks by malaise, fever, cephalalgia, stiff neck, myalgias,arthralgia, or lymphadenopathy. The EM constitutes the first stage or phase of thedisease <strong>and</strong> lasts a few weeks, but may recur. In the second stage, after several weeksor months have passed <strong>and</strong> the agent has disseminated, some patients develop multipleEMs, meningoencephalitis, neuropathies, myocarditis, <strong>and</strong> atrioventriculartachycardia. Some suffer arthritic attacks in the large joints, which may recur forseveral years, at times taking a chronic course (Steere et al., 1983). Months or yearslater, the third stage may occur in some patients; this stage sometimes includes acrodermatitischronica atrophicans <strong>and</strong> neurological <strong>and</strong> articular changes.


LYME DISEASE 181It should be borne in mind that the connection between EM <strong>and</strong> arthritis might notbe apparent, as several weeks or months transpire between the two episodes. Of 405patients showing EM, 249 had later neurological, cardiac, <strong>and</strong> articular symp<strong>to</strong>ms(Steere <strong>and</strong> Malawista, 1979). In Europe, cases with arthritis are rare, while neurologicalsymp<strong>to</strong>ms <strong>and</strong> acrodermatitis are more frequent.Treatment for Lyme disease consists of giving the patient doxycycline for 10 <strong>to</strong> 30days, or ceftriaxone, particularly if there is a neurological disorder (Benenson, 1990).The Disease in Animals: The effect of the spirochete infection on wild animalsis not known, but it may be asymp<strong>to</strong>matic. The predominant symp<strong>to</strong>m in dogs islameness due <strong>to</strong> arthritis in different joints, which may be migra<strong>to</strong>ry. Arthralgia isoften accompanied by fever, anorexia, fatigue, <strong>and</strong> lymphadenitis. Arthritis is usuallytemporary, but may become chronic.Different symp<strong>to</strong>ms have been observed in horses, including arthritis, encephalitis,uveitis, dermatitis, edema of the limbs, <strong>and</strong> death of colts associated with naturalinfection in pregnant mares. However, the infection has not been confirmed inany of the cases described (Cohen et al., 1992).In cattle, infection caused by B. burgdorferi has also been associated with lameness.Serological analysis using Western blot, ELISA, <strong>and</strong> indirect immunofluorescencetechniques on 27 milk cows from 17 herds in Minnesota <strong>and</strong> Wisconsin foundthat high serological titers were associated with arthritis (Wells et al., 1993).Source of Infection <strong>and</strong> Mode of Transmission: The etiologic agent is transmittedby a vec<strong>to</strong>r, which in the US is the tick Ixodes dammini on the NortheastCoast <strong>and</strong> northern states of the Midwest, but I. pacificus on the West Coast(Benenson, 1990). The vec<strong>to</strong>r is I. ricinus in Europe, possibly I. holocyclus inAustralia (Stewart et al., 1982), <strong>and</strong> I. persulcatus in Asia.Isolation of the etiologic agent has made it possible <strong>to</strong> definitively establish therole of ticks as vec<strong>to</strong>rs. In fact, in the endemic area of Connecticut, a spirochetewith the same antigenic <strong>and</strong> morphological characteristics as the one in Lyme diseasepatients was isolated from 21 (19%) of 110 nymphs <strong>and</strong> adult ticks (I.dammini). The high rate of infection of the vec<strong>to</strong>r was shown by direct immunofluorescence;in one locality, 30 (21%) of 143 I. dammini contained spirochetes,<strong>and</strong> in another, 17 (26%) of 66 contained spirochetes. These results were obtainedonly for nymphs <strong>and</strong> adults that had fed, while 148 larvae that had not fed werenegative (Steere et al., 1983). On Shelter Isl<strong>and</strong>, New York, more than 50% of tickswere infected (Bosler et al., 1983). In contrast, only 2% of the I. pacificus tickswere infected.The fact that the larvae were not infected prior <strong>to</strong> feeding on blood would indicatethat the tick becomes infected from an animal reservoir. This reservoir would besmall rodents <strong>and</strong> other wild mammals; among these the white-footed mouse(Peromyscus leucopus) is considered very important on the East Coast of the US. InEurope, the reservoir of the infection is also small, wild rodents, such as Apodemussylvaticus <strong>and</strong> Clethrionomys spp. Tick larvae <strong>and</strong> nymphs feed on the blood ofthese small mammals <strong>and</strong> become infected with B. burgdorferi. The adult tick maytransmit the etiologic agent <strong>to</strong> a very small percentage of the eggs, but there is agradual loss of the agent when they go on <strong>to</strong> the larval <strong>and</strong> nymph stages until it disappearscompletely. The infection is renewed when larval <strong>and</strong> nymph ticks feed onrodents (Burgdorfer et al., 1989). This fact is reflected in the high percentage of lar-


182 BACTERIOSESvae <strong>and</strong> nymphs found on these small wild mammals, as well as their high rate ofinfection by B. burgdorferi. The adult tick has a predilection for deer (Odocoileusvirginianus) in the foci along the eastern coast of the US. This cycle is repeated inother areas of the world, with different animal species whose blood feeds the stagesof various tick species. The bio<strong>to</strong>pe where these cycles develop is wooded areas orareas of dense vegetation that retain the moisture that is favorable <strong>to</strong> ticks (Madigan<strong>and</strong> Tleitler, 1988).Adult ticks are abundant in spring <strong>and</strong> fall; nymphs, in spring <strong>and</strong> early summer;<strong>and</strong> larvae, in late summer <strong>and</strong> early fall. All stages in the development of ticks areparasitic in humans, but the nymph stage is primarily responsible for the transmissionof B. burgdorferi <strong>to</strong> man (Anderson, 1989; Steere, 1990).Role of Animals in the Epidemiology of the Disease: On the basis of currentinformation, it can be asserted that wild animals are primarily responsible for maintainingthe infection in natural foci. Dogs <strong>and</strong> birds may spread ticks <strong>and</strong> increaseendemic areas. Man is an accidental host.Diagnosis: Until recently, diagnosis was based exclusively on the clinical picture,especially a his<strong>to</strong>ry of EM, <strong>and</strong> on epidemiological information.Although now possible, isolation of the infective agent by culture is still not verypractical. In 1983, Steere et al. isolated the agent in only three patients, using a <strong>to</strong>talof 142 clinical samples taken from 56 patients. Barbour, S<strong>to</strong>ener, Kelly (BSK)medium is used for isolation <strong>and</strong> is incubated at 33°C; it is easier <strong>to</strong> isolate the agentfrom cutaneous lesions than from blood. The indirect immunofluorescence test withconjugated IgM <strong>and</strong> IgG sera was widely used. Patients with EM had elevated IgMantibody titers only between the EM phase <strong>and</strong> convalescence two <strong>to</strong> three weekslater. Patients with late manifestations of the disease (arthritis, cardiac, or neurologicalanomalies) had elevated IgG antibody titers (Steere et al., 1983). It was latershown that indirect ELISA was more sensitive <strong>and</strong> specific than immunofluorescence(Steere, 1990). In serological tests, there may be cross-reactions with otherspirochetes. Given that all serological tests have limited specificity <strong>and</strong> sensibility,their use is not recommended for asymp<strong>to</strong>matic individuals.Diagnosis in animals is similar <strong>to</strong> that in humans. Early treatment with antibioticsshortens the duration of EM <strong>and</strong> may prevent or lessen late manifestations of the disease;it may also have an effect on reducing the level of antibodies.Control: The only methods of prevention consist of avoiding endemic areas <strong>and</strong>tick bites. Persons entering natural foci should use protective footwear <strong>and</strong> clothing,though this is not always possible. Insect repellents may provide some protection. Itis advisable <strong>to</strong> check the body frequently <strong>and</strong> remove attached ticks by pulling gentlywith tweezers pressed as closely as possible <strong>to</strong> the skin. It is recommended thatgloves be used during this operation.Dogs should be checked frequently <strong>and</strong> ticks should be removed as carefully aswith humans. The use of tickicides in powder form or collars is a good preventivemeasure. There is currently an inactivated commercial vaccine available for dogs. Itis administered in two doses at three week intervals <strong>and</strong> annually thereafter (Chu etal., 1992). Widespread <strong>and</strong> indiscriminate use of this vaccine is a matter of discussion,although it is recognized that the bacterin has no side effects (Kazmierczak <strong>and</strong>Sorhage, 1993).


LYME DISEASE 183BibliographyAnderson, J.F. Epizootiology of Borrelia in Ixodes tick vec<strong>to</strong>rs <strong>and</strong> reservoir hosts. RevInfect Dis 11(Suppl):1451–1459, 1989.Anderson, J.F., L.A. Magnarelli. Spirochetes in Ixodes dammini <strong>and</strong> Babesia microti onPrudence Isl<strong>and</strong>, Rhode Isl<strong>and</strong>. J Infect Dis 148:1124, 1983.Anderson, J.F., L.A. Magnarelli. Avian <strong>and</strong> mammalian hosts for spirochete-infected ticks<strong>and</strong> insects in a Lyme disease focus in Connecticut. Yale J Biol Med 57:627–641, 1984.Barbour, A.G. Isolation <strong>and</strong> cultivation of Lyme disease spirochetes. Yale J Biol Med57:521–525, 1984.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bernard, W.V., D. Cohen, E.M. Bosler, D. Zamos. Serologic survey for Borrelia burgdorferiantibody in horses referred <strong>to</strong> a mid-Atlantic veterinary teaching hospital. J Am Vet MedAssoc 196:1255–1258, 1990.Bosler, E.M., J.L. Coleman, J.L. Benach, et al. Natural distribution of the Ixodes damminispirochete. Science 220:321–322, 1983.Bosler, E.M., B.G. Ormis<strong>to</strong>n, J.L. Coleman, et al. Prevalence of the Lyme disease spirochetein populations of white-tailed deer <strong>and</strong> white-footed mice. Yale J Biol Med 57:651–659, 1984.Bruhn, F.W. Lyme disease. Am J Dis Child 138:467–470, 1984.Burgdorfer, W., A.G. Barbour, S.F. Hayes, et al. Lyme disease—A tickborne spiroche<strong>to</strong>sis?Science 216:1317–1319, 1982.Burgdorfer, W., S.F. Hayes, D. Corwin. Pathophysiology of the Lyme disease spirochete,Borrelia burgdorferi, in ixodid ticks. Rev Infect Dis 11(Suppl 6):1442–1450, 1989.Burgess, E.C. Natural exposure of Wisconsin dogs <strong>to</strong> the Lyme disease spirochete (Borreliaburgdorferi). Lab Anim Sci 36:288–290, 1986.Chu, H.J., L.G. Chavez, B.M. Blumer, et al. Immunogenicity <strong>and</strong> efficacy study of a commercialBorrelia burgdorferi bacterin. J Am Vet Med Assoc 201:403–411, 1992.Cohen, N.D., C.N. Carter, M.A. Thomas, Jr., et al. Clinical <strong>and</strong> epizootiologic characteristicsof dogs seropositive for Borrelia burgdorferi in Texas: 110 cases (1988). J Am Vet MedAssoc 197:893–898, 1990.Cohen, N.D., F.C. Heck, B. Heim, et al. Seroprevalence of antibodies <strong>to</strong> Borrelia burgdorferiin a population of horses in central Texas. J Am Vet Med Assoc 201:1030–1034, 1992.Charmot, G., F. Rodhain, C. Perez. Un cas d’arthrite de Lyme observé en France. NouvPresse Med 11:207–208, 1982.Gerster, J.C., S. Guggi, H. Perroud, R. Bovet. Lyme arthritis appearing outside the UnitedStates: A case report from Switzerl<strong>and</strong>. Brit Med J 283:951–952, 1981.Hanrahan, J.P., J.L. Benach, J.L. Coleman, et al. Incidence <strong>and</strong> cumulative frequency ofendemic Lyme disease in a community. J Infect Dis 150:489–496, 1984.Hayes, S.F., W. Burgdorfer, A.G. Barbour. Bacteriophage in Ixodes dammini spirochete, etiologicalagent of Lyme disease. J Bacteriol 154:1436–1439, 1983.Johnson, R.C., F.W. Hyde, C.M. Rumpel. Taxonomy of the Lyme disease spirochetes. YaleJ Biol Med 57:529–537, 1984.Johnson, R.C., F.W. Hyde, A.G. Steigerwalt, D.J. Brenner. Borrelia burgdorferi sp. nov.:Etiologic agent of Lyme disease. Int J Syst Bacteriol 34:496–497, 1984.Kazmierczak, J.J., F.E. Sorhage. Current underst<strong>and</strong>ing of Borrelia burgdorferi infection,with emphasis on its prevention in dogs. J Am Vet Med Assoc 203:1524–1528, 1993.Madigan, J.E., J. Teitler. Borrelia burgdorferi borreliosis. J Am Vet Med Assoc192:892–896, 1988.Magnarelli, L.A., J.F. Anderson, W. Burgdorfer, W.A. Chappel. Parasitism by Ixodesdammini (Acari: Ixodidae) <strong>and</strong> antibodies <strong>to</strong> spirochetes in mammals at Lyme disease foci inConnecticut, USA. J Med En<strong>to</strong>mol 21:52–57, 1984.


184 BACTERIOSESOliver, J.N., M.R. Owsley, H.J. Hutcheson, et al. Conspecificity of the ticks Ixodes scapularis<strong>and</strong> I. dammini (Acari: Ixodidae). J Med En<strong>to</strong>mol 30:54–63, 1993.Russell, H., J.S. Sampson, G.P. Schmid, et al. Enzyme-linked immunosorbent assay <strong>and</strong>indirect immunofluorescence assay for Lyme disease. J Infect Dis 149:465–470, 1984.Schmid, G.P. The global distribution of Lyme disease. Rev Infect Dis 7:41–50, 1985.Schulze, T., G.S. Bowen, E.M. Bosler, et al. Amblyomma americanum:A potential vec<strong>to</strong>rof Lyme disease in New Jersey. Science 224:601–603, 1984.Stanek, G., G. Wewalka, V. Groh, et al. Differences between Lyme disease <strong>and</strong> Europeanarthropod-borne Borrelia infections. Lancet 1(8425):401, 1985.Steere, A.C. Borrelia burgdorferi (Lyme disease, Lyme borreliosis). In: M<strong>and</strong>ell, G.L., R.G.Douglas, Jr., J.E. Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. NewYork: Churchill Livings<strong>to</strong>ne, Inc.; 1990.Steere, A.C., J. Green, R.T. Schoen. Successful parenteral penicillin therapy of establishedLyme arthritis. N Engl J Med 312:869–874, 1985.Steere, A.C., R.L. Grodzicki, A.N. Kornblatt, et al. The spirochetal etiology of Lyme disease.N Engl J Med 308:733–744, 1983.Steere, A.C., S.E. Malawista. Cases of Lyme disease in the United States: Locations correlatedwith distribution of Ixodes dammini. Ann Intern Med 91:730–733, 1979.Stevens, R., K. Hechemy, A. Rogers, J. Benach. Fluoroimmunoassay (FIAX) for Lyme diseaseantibody. Abstracts, Annual Meeting of the American Society for Microbiology, March3–7, 1985.Stewart, A., J. Glass, A. Patel, G. Watt, A. Cripps, R. Clancy. Lyme arthritis in the HunterValley. Med J Aust 1:139, 1982.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Current Trends Update: Lyme disease <strong>and</strong> cases occurringduring pregnancy—United States. MMWR Morb Mortal Wkly Rep 34:376–378, 383–384, 1985.Wells, S.J., M. Trent, R.A. Robinson, et al. Association between clinical lameness <strong>and</strong>Borrelia burgdorferi antibody in dairy cows. Am J Vet Res 54:398–405, 1993.White, D.J., H.G. Chang, J.L. Benach, et al. The geographic spread <strong>and</strong> temporal increaseof the Lyme disease epidemic. JAMA 266:1230–1236, 1991.Wilkinson, H.W. Immunodiagnostic tests for Lyme disease. Yale J Biol Med 57:567–572, 1984.


MELIOIDOSISICD-10 A24.1 acute <strong>and</strong> fulminating melioidosis;A24.2 subacute <strong>and</strong> chronic melioidosis; A24.3 other melioidosisSynonyms: Whitmore’s disease, rodent gl<strong>and</strong>ers.Etiology: Pseudomonas (Malleomyces) pseudomallei, a small, aerobic, motile,gram-negative bacillus closely related <strong>to</strong> P. mallei, the agent of gl<strong>and</strong>ers. Whenstained with methylene blue or Wright stain, it shows bipolar coloration, in the shapeof a safety pin. It is pleomorphic <strong>and</strong> sometimes forms chains. It is a saprophytic


MELIOIDOSIS 185bacteria that lives in surface waters <strong>and</strong> in soil. P. pseudomallei can survive in moist,clayey soil; under labora<strong>to</strong>ry conditions; at ambient temperature; <strong>and</strong> in shade for 30months (Thomas <strong>and</strong> Forbes-Faulkner, 1981).P. pseudomallei has several possible virulence fac<strong>to</strong>rs, including an endo<strong>to</strong>xin, anexo<strong>to</strong>xin, <strong>and</strong> various digestive enzymes that can attack tissue. The role that each ofthese virulence fac<strong>to</strong>rs plays in pathogenesis is still unknown. The exo<strong>to</strong>xin is themost <strong>to</strong>xic substance produced by the bacteria <strong>and</strong> can inhibit intracellular proteinsynthesis (Dance, 1991; Ismail et al., 1991).Geographic Distribution: Most human <strong>and</strong> animal cases have been recorded inSoutheast Asia (Indonesia, Malaysia, Myanmar [Burma], <strong>and</strong> Thail<strong>and</strong>), which isconsidered the main endemic area. The disease has also been diagnosed in northeasternAustralia, Guam, Iran, Korea, Madagascar, Papua New Guinea, Sri Lanka,<strong>and</strong> Turkey. Cases have also occurred in Bangladesh, India, <strong>and</strong> Pakistan. In theAmericas, the infection has been confirmed in Aruba, the Bahamas, Ecuador, ElSalvador, Mexico, Panama, <strong>and</strong> Puer<strong>to</strong> Rico. More recent investigations haverevealed the agent’s presence in other areas (Brazil, Burkina Faso, Côte d’Ivoire,Haiti, <strong>and</strong> Peru) by isolating it from people, animals, or soil <strong>and</strong> water samples.Sporadic cases have also occurred in human in Kenya <strong>and</strong> the Gambia, in swine inBurkina Faso <strong>and</strong> Niger, <strong>and</strong> in goats in Chad. The agent’s distribution is predominantlytropical. The epizootic that occurred in the “Jardin des Plantes,” in Paris, isthe first reported outbreak in a temperate climate. In Europe, in addition <strong>to</strong> France,there have been cases in horses in Spain (Benenson, 1990; Galimard <strong>and</strong> Dodin,1982; Dance, 1991a).Occurrence in Man: Clinically apparent infection caused by P. pseudomallei isnot very <strong>common</strong>. During the war in Indochina, several hundred French, American,<strong>and</strong> Vietnamese soldiers became ill with melioidosis. From 1965 <strong>to</strong> 1969, threecases per month occurred among US Army soldiers in Vietnam (Piggott, 1976).According <strong>to</strong> a serological survey, 9% of the 3 million US personnel participating inthe Vietnam conflict were exposed <strong>to</strong> the agent. Cases confirmed in the US duringthe 1970s were almost all military personnel or travelers returning from SoutheastAsia (CDC, 1977).The numerous cases that occurred among military personnel during the VietnamWar provoked interest in the disease among medical professionals in Thail<strong>and</strong>. Prior<strong>to</strong> 1965, only three cases of melioidosis had been recorded in that country, whilethere were a <strong>to</strong>tal of about 1,000 cases between 1967 <strong>and</strong> 1988 (Kanai <strong>and</strong>Dejsirilert, 1988).Melioidosis is currently recognized as the most <strong>common</strong> cause of pneumoniaoccurring in the Top End region in the Northern Terri<strong>to</strong>ry of Australia(Currie, 1993).Occurrence in Animals: In endemic zones, sporadic cases have been reported indifferent animal species. Occasional outbreaks have occurred among sheep (inAustralia <strong>and</strong> Aruba), in swine (Vietnam), in goats, cattle, horses, dogs, dolphins,tropical fish, <strong>and</strong> zoo animals, as well as in monkeys imported for labora<strong>to</strong>ries. Acase occurred in macaques of the species Macaca fascicularis imported <strong>to</strong> GreatBritain from the Philippines. There were a <strong>to</strong>tal of 13 confirmed or suspected casesin 50 imported animals. Most had splenic abscesses, but their general condition was


186 BACTERIOSESnot affected <strong>and</strong> infection was suspected on the basis of results from serological tests(Dance et al., 1992).The Disease in Man: The incubation period may be a few days, but in somepatients the agent lies dormant for months, or even years, before clinical signs areseen. The infection may occur subclinically, as was shown by a serologic survey ofwar veterans, or the disease may take an acute <strong>and</strong> fulminant, or subacute <strong>and</strong>chronic form. In the acute form, the patient dies in a few days, after suffering fever,pneumonia, <strong>and</strong> gastroenteritis. The disease generally appears as a respira<strong>to</strong>ry illnessthat varies from mild bronchitis <strong>to</strong> severe <strong>and</strong> fatal pneumonia. Case mortalityis approximately 30% (Kanai <strong>and</strong> Dejsirilert, 1988).In septicemic cases of short duration, the principal lesion consists of smallabscesses distributed throughout the body. When septicemia is prolonged, larger,confluent abscesses are found, often localized in one organ.Lasting from a few months <strong>to</strong> many years, the subacute <strong>and</strong> chronic form is characterizedby localization in some organ, such as the lungs, lymph gl<strong>and</strong>s, skin, orbones. The lesion consists of a combination of necrosis <strong>and</strong> granuloma<strong>to</strong>us inflammation.The central zone of necrosis contains a purulent or caseous exudate that canbe confused with a tubercular lesion.In endemic areas, such as Southeast Asia, seroepidemiological surveys indicatethat latent or subclinical forms are <strong>common</strong>. In latent cases, P. pseudomallei mayremain inactive for years <strong>and</strong> become activated when there is some other disease oran individual’s defenses are lowered due <strong>to</strong> the administration of steroids or otherimmunosuppressant therapy (Kanai <strong>and</strong> Dejsirilert, 1988).In northern Australian aborigines, a form of the disease has been observed inwhich primary localization is in the lower urogenital system. This localization wasobserved in 6 of 16 aborigines with melioidosis (Webling, 1980).Although the infection may occur in healthy individuals, P. pseudomallei islargely an opportunistic bacteria. In Thail<strong>and</strong>, 70% of patients have some concurrentdisease, particularly diabetes or renal deficiency (Dance, 1991b). The ratio betweenmen <strong>and</strong> women affected is 3:2. Most cases occur during or after tropical rains.Various beta-lactamic agents are bactericides for P. pseudomallei. One of thesecompounds, ceftazidime, reduced mortality by 50% among individuals with acute<strong>and</strong> severe melioidosis (White et al., 1989).The Disease in Animals: Many animal species are susceptible. Sporadic caseshave been observed in sheep, goats, horses, swine, cattle, dogs, cats, nonhuman primates,wild <strong>and</strong> peridomestic rats, other wild animals, labora<strong>to</strong>ry guinea pigs, <strong>and</strong>rabbits. The most susceptible species are sheep, swine, <strong>and</strong> goats, in which epidemicoutbreaks have occurred.As seen in Aruba, the disease in sheep consisted primarily of abscesses of the viscera,joints, <strong>and</strong> lymph nodes. In a few weeks, 25 of 90 sheep died from the disease<strong>and</strong> many survivors suffered weight loss <strong>and</strong> polyarthritis (Sutmöller et al., 1957).In cases in Australian sheep, cough <strong>and</strong> nervous symp<strong>to</strong>ms were also observed(Laws <strong>and</strong> Hall, 1964).In swine, the symp<strong>to</strong>ma<strong>to</strong>logy consists of fever, prostration, dyspnea, cough, <strong>and</strong>arthritis. In suckling pigs, the disease is often fatal. In addition, the disease may befound through necropsy or when meat is seized in slaughterhouses. In northernQueensl<strong>and</strong> (Australia), melioidosis occurs sporadically in swine bred in contact


MELIOIDOSIS 187with the soil. In the southern part of the same state, which is not an enzootic area,veterinary inspection at a slaughterhouse uncovered cases in suckling pigs fromeight intensive breeding facilities, during three successive years. These cases (159out of 17,397 animals inspected) occurred after abundant rains <strong>and</strong> flooding.Abscesses were found in the bronchial ganglia of 40% <strong>and</strong> in the spleen of 34%. Theoutbreak was attributed <strong>to</strong> inhalation of aerosols from water (Ketterer et al., 1986).The disease is rare in cattle. The etiologic agent has been isolated from splenicabscesses, from the central nervous system, <strong>and</strong> from aborted fetuses.In horses, the infection may become apparent due <strong>to</strong> the symp<strong>to</strong>ms of septicemia,colic, diarrhea, <strong>and</strong> edemas in the legs.The symp<strong>to</strong>ma<strong>to</strong>logy is not very characteristic <strong>and</strong> the disease is difficult <strong>to</strong> diagnosein animal species in which it occurs sporadically. The lesions, which are similar<strong>to</strong> those in man, may suggest melioidosis <strong>and</strong> lead <strong>to</strong> its diagnosis.Source of Infection <strong>and</strong> Mode of Transmission (Figure 13): Investigations haveshown that the reservoirs of P. pseudomallei are surface waters <strong>and</strong> soil, as corroboratedby sampling done in Southeast Asia. The highest isolation rates wereobtained in rice fields <strong>and</strong> newly planted oil palm plantations (14.5%–33.3% of theisolations were from water samples). Seroepidemiologic studies also show that thehighest reac<strong>to</strong>r rates <strong>to</strong> the hemagglutination test came from workers or inhabitantsof those areas. Human <strong>and</strong> animal infection occurs mainly during the rainy season.The etiologic agent can survive for many months in surface water <strong>and</strong>, with its lownutritional requirements, it can multiply in the hot, humid environment characteristicof endemic regions.Figure 13. Melioidosis (Pseudomonas pseudomallei). Mode of transmission.Contaminated surfacewaters <strong>and</strong> soilManSheep, goats,horses, swine,cattle, dogs, cats,nonhumanprimates,wild <strong>and</strong>semi-domesticrats


188 BACTERIOSESTransmission from animal <strong>to</strong> animal or from animal <strong>to</strong> man has not been proven,but it is thought that in some cases the infection may be passed from person <strong>to</strong> person.In addition <strong>to</strong> a case in Vietnam in which an American soldier with prostatitisseems <strong>to</strong> have transmitted the disease venereally <strong>to</strong> a woman, venereal transmissionof the disease was also suspected among Australian aborigines with urogenitalmelioidosis. In accordance with tribal rituals, these aborigines smear their genitalswith clay <strong>and</strong> coitus normally takes place in contact with the soil (Webling, 1980).It is accepted that humans <strong>and</strong> animals acquire the infection through contact withcontaminated water or soil, primarily through skin abrasions, but also throughinhalation of dust <strong>and</strong> ingestion of contaminated water. During the war in Indochina,the number of recorded human cases climbed considerably due <strong>to</strong> contaminationof war wounds with mud, traversal of flooded countryside, or prolonged stayin trenches.The rat flea Xenopsylla cheopis <strong>and</strong> the mosqui<strong>to</strong> Aedes aegypti are capable oftransmitting the infection experimentally <strong>to</strong> labora<strong>to</strong>ry animals. The etiologic agentmultiplies in the digestive tract of these insects. The role of these possible vec<strong>to</strong>rsin natural transmission has not yet been evaluated, but it is thought <strong>to</strong> be oflittle importance.Role of Animals in the Epidemiology of the Disease: Melioidosis seems <strong>to</strong> bea disease <strong>common</strong> <strong>to</strong> man <strong>and</strong> animals, with water <strong>and</strong> soil as reservoirs <strong>and</strong> sourcesof infection for both. Nevertheless, animals are believed <strong>to</strong> play a role as hosts intransporting the etiologic agent <strong>to</strong> new geographic areas.Diagnosis: The only incontrovertible diagnostic method is isolation <strong>and</strong>identification of the etiologic agent, by either direct culture or inoculation of guineapigs. P. pseudomallei can be isolated from abscesses, sputum, blood, urine, <strong>and</strong>various tissues.The allergenic test using melioidin may be useful for diagnosis in animals, butgives many false negatives in swine <strong>and</strong> false positives in goats.Of the serologic tests, indirect hemagglutination with melioidin-sensitized erythrocyteshas proven <strong>to</strong> be sufficiently sensitive <strong>and</strong> specific in nonendemic areas.In endemic areas, titers of ≥ 1:160 would have <strong>to</strong> be considered significant reactions(Appassakij et al., 1990). The indirect immunofluorescence <strong>and</strong> ELISA testshave proven <strong>to</strong> be more sensitive <strong>and</strong> specific (Dance, 1991). A latex agglutinationtest developed <strong>and</strong> evaluated by Smith et al. (1993) is considered highly sensitive<strong>and</strong> specific.The ELISA test was used on Malaysian sheep <strong>to</strong> detect the anti-exo<strong>to</strong>xin. Theanti<strong>to</strong>xin was confirmed in 49.3% of the sera taken from a sheep herd that had beennaturally exposed <strong>to</strong> the infection. In sheep kept on a property without infection, therate was 6% (Ismail et al., 1991).Control: Since it is an infrequent disease, specific preventive measures are notjustified. In man, the use of boots during outdoor work can provide a certain amoun<strong>to</strong>f protection against the infection in endemic areas. Proper treatment of wounds <strong>and</strong>abrasions is important.In animals, control of the infection is difficult, unless the environment is changedthrough such measures as drainage of low-lying, flooded fields.


MELIOIDOSIS 189BibliographyAppassakij, H., K.R. Silpapojakul, R. Wansit, M. Pornpatkul. Diagnostic value of the indirecthemagglutination test for melioidosis in an endemic area. Am J Trop Med Hyg42:248–253, 1990.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Biegeleisen, J.Z., Jr., M.R. Mosquera, W.B. Cherry. A case of human melioidosis. Clinical,epidemiological <strong>and</strong> labora<strong>to</strong>ry findings. Am J Trop Med Hyg 13:89–99, 1966.Currie, B. Medicine in tropical Australia. Med J Aust 158:609, 612–615, 1993.Dance, D.A. Pseudomonas pseudomallei: Danger in the paddy fields. Trans R Soc TropMed Hyg 85:1–3, 1991a.Dance, D.A. Melioidosis: The tip of the iceberg? Clin Microbiol Rev 4:52–60, 1991b.Dance, D.A., C. King, H. Aucken, et al. An outbreak of melioidosis in imported primatesin Britain. Vet Rec 130:525–529, 1992.Galimard, M., A. Dodin. Le point sur la melioidose dans le monde. Bull Soc Pathol Exot75:375–383, 1982.Hubbert, W.T. Melioidosis: Sources <strong>and</strong> potential. Wildl Dis 5(3):208–212, 1969.Ismail, G., R. Mohamed, S. Rohana, et al. Antibody <strong>to</strong> Pseudomonas pseudomallei exo<strong>to</strong>xinin sheep exposed <strong>to</strong> natural infection. Vet Microbiol 27:277–282, 1991.Kanai, K., S. Dejsirilert. Pseudomonas pseudomallei <strong>and</strong> melioidosis, with special reference<strong>to</strong> the status in Thail<strong>and</strong>. Jpn J Med Sci Biol 41:123–157, 1988.Kaufman, A.E., A.D. Alex<strong>and</strong>er, A.M. Allen, R.J. Cronkin, L.A. Dillingham, J.D. Douglas,et al. Melioidosis in imported nonhuman primates. Wildl Dis 6:211–219, 1970.Ketterer, P.J., B. Donald, R.J. Rogers. Bovine melioidosis in south-eastern Queensl<strong>and</strong>.Aust Vet J 51:395–398, 1975.Ketterer, P.J., W.R. Webster, J. Shield, et al. Melioidosis in intensive piggeries in south-easternQueensl<strong>and</strong>. Aust Vet J 63:146–149, 1986.Laws, L., W.T.K. Hall. Melioidosis in animals in north Queensl<strong>and</strong>. IV. Epidemiology. AustVet J 40:309–314, 1964.Piggott, J.A. Melioidosis. In: Binford, C.H., D.H. Connor, eds. Pathology of Tropical <strong>and</strong>Extraordinary Diseases. Washing<strong>to</strong>n, D.C.: Armed Forces Institute of Pathology; 1976.Piggott, J.A., L. Hochholzer. Human melioidosis. A his<strong>to</strong>pathologic study of acute <strong>and</strong>chronic melioidosis. Arch Pathol 90:101–111, 1970.Redfearn, M.S., N.J. Palleroni. Gl<strong>and</strong>ers <strong>and</strong> melioidosis. In: Hubbert, W.T., W.F.McCulloch, P.R. Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed.Springfield: Thomas; 1975.Smith, M.D., V. Wuthiekanun, A.L. Walsh, T.L. Pitt. Latex agglutination test for identificationof Pseudomonas pseudomallei. J Clin Pathol 46:374–375, 1993.Strauss, J.M., M.G. Groves, M. Mariappan, D.W. Ellison. Melioidosis in Malaysia. II.Distribution of Pseudomonas pseudomallei in soil <strong>and</strong> surface water. Am J Trop Med Hyg18:698–702, 1969.Sutmöller, P., F.C. Kraneveld, A. Van der Schaaf. Melioidosis (Pseudomalleus) in sheep,goats, <strong>and</strong> pigs in Aruba (Netherl<strong>and</strong>s Antilles). J Am Vet Med Assoc 130:415–417, 1957.Thomas, A.D., J.C. Forbes-Faulkner. Persistence of Pseudomonas pseudomallei in soil.Aust Vet J 57:535–536, 1981.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Melioidosis—Pennsylvania. MMWR Morb Mortal Wkly Rep25:419–420, 1977.Webling, D.D. Geni<strong>to</strong>-urinary infections with Pseudomonas pseudomallei in Australianaboriginals. Trans R Soc Trop Med Hyg 74:138–139, 1980.


190 BACTERIOSESWhite, N.J., D.A. Dance, W. Chaowagul, et al. Halving of mortality of severe melioidosisby ceftazidime. Lancet 2:697–701, 1989.


NECROBACILLOSISICD-10 A48.8 other specified bacterial <strong>diseases</strong>Synonyms: Schmorl’s disease, calf diphtheria, foot rot.Etiology: Fusobacterium necrophorum, a nonsporulating, obligate anaerobe thatis a pleomorphic, gram-negative bacillus of the family Bacteroidaceae. In broth cultures,F. necrophorum varies from coccoid shapes <strong>to</strong> filaments with granular inclusions.Rod shapes are more <strong>common</strong> in agar cultures. This bacteria is a componen<strong>to</strong>f the normal flora of the mouth, gastrointestinal tract, <strong>and</strong> urogenital tract of man<strong>and</strong> animals. Strains have varied virulence categories: pathogenic for mice; slightlypathogenic or not at all, but hemolytic, like the first category; <strong>and</strong> a third category(formerly called Sphaerophorus pseudonecrophorus) that is neither hemolytic norpathogenic. There may be mutation from one category (or phase) <strong>to</strong> another. Thevalidity of the identification of this bacteria in works prior <strong>to</strong> 1970 is questioned(Holdeman et al., 1984).Different species of Bacteroides play an important pathogenic role in necrobacillosis.They may appear alone or in conjunction with other species of the same genus,particularly in man, or with F. necrophorus in animals. Bacteroides spp. is also anonsporulating, gram-negative, obligate anaerobe. Bacteroides nodosus is of particularinterest in sheep pathology. These bacteria are nonmotile, <strong>and</strong> take the shape ofstraight or slightly curved rods sized 1 <strong>to</strong> 1.7 by 3 <strong>to</strong> 6 microns. They appear singlyor in pairs <strong>and</strong> often have thickened ends (Holdeman et al., 1984). They have numerouspili (fimbriae), an important virulence fac<strong>to</strong>r. The pili likely play an importantrole in colonization of the epidermal matrix of hooves. These appendices also makeit possible <strong>to</strong> sub-classify the agent serologically in<strong>to</strong> 9 serogroups containing 16 <strong>to</strong>20 serovars or serotypes, according <strong>to</strong> their determination in different countries(Gradin et al., 1991).The polymicrobial nature of most anaerobic infections in man makes it difficult<strong>to</strong> distinguish the true pathogen or pathogens from those that merely accompany theinfection (Kirby et al., 1980). Singly or acting jointly with other nonsporulating,anaerobic bacteria, F. necrophorum causes different <strong>diseases</strong> <strong>and</strong> pathological conditionsin man <strong>and</strong> animals. Different species of the genus Bacteroides, whichbelong <strong>to</strong> the same family as F. necrophorum, cause disease either by themselves (inman) or in combination <strong>and</strong> at times in synergistic action with F. necrophorum (inman <strong>and</strong> animals).Geographic Distribution: Worldwide.


NECROBACILLOSIS 191Occurrence in Man: Advances in labora<strong>to</strong>ry technology for the isolation ofanaerobes have led <strong>to</strong> greater recognition of their role in human pathology <strong>and</strong>, consequently,<strong>to</strong> an increase in the number of recorded cases in medical facilities.Occurrence in Animals: Some <strong>diseases</strong>, such as foot rot in sheep, occur frequentlyin all countries where sheep are raised. Others, such as calf diphtheria(necrobacillary s<strong>to</strong>matitis), are less <strong>common</strong>. Bovine hepatic necrobacillosis causesappreciable economic losses in many countries due <strong>to</strong> confiscation of animals inslaughterhouses; it is more frequent in areas where cattle are fed grain in feedlots(Timoney et al., 1988).The Disease in Man: F. necrophorum causes a wide variety of necrotic lesions,empyema, pulmonary abscesses, arthritis, <strong>and</strong> ovariosalpingitic sepsis. Bacteroidesfragilis <strong>and</strong> F. necrophorum are important agents of cerebral abscesses <strong>and</strong>, occasionally,of meningitis, almost always as a consequence of an otitis media (Islam <strong>and</strong>Shneerson, 1980). The formerly high incidence of septicemia caused by F.necrophorum in children <strong>and</strong> adolescents who had suffered from <strong>to</strong>nsillitis(Lemierre’s syndrome) has now diminished notably <strong>and</strong> constitutes only 1% <strong>to</strong> 2%of all bacteremias caused by anaerobes. Patients with septicemia usually exhibitexudative pharyngitis or a peri<strong>to</strong>nsillar abscess, but these symp<strong>to</strong>ms may disappearby the time some patients obtain medical attention (Seidenfeld et al., 1982). In mosthuman clinical specimens, only the genera Bacteroides, Prevotella, Porphyromonas,<strong>and</strong> Fusobacterium should be considered among the anaerobic bacilli (Jousimies-Somer <strong>and</strong> Finegold, 1991). Infections in man come from the normal flora of adjacentcavities.The most effective antibiotics for treating infections caused by gram-negativeanaerobes are metronidazole, chloramphenicol, <strong>and</strong> imipenem (Jousimies-Somer<strong>and</strong> Finegold, 1991).The Disease in Animals: F. necrophorum is more important in animal than inhuman pathology <strong>and</strong> is the cause of several <strong>common</strong> <strong>diseases</strong>.SHEEP: Foot rot is the most <strong>common</strong> cause of lameness in sheep. The diseasebegins with interdigital dermatitis, progresses <strong>to</strong> the epidermal matrix of the hooves,<strong>and</strong> then causes destruction of the interdigital skin <strong>and</strong> detachment of the hoof.Environmental fac<strong>to</strong>rs, such as wet soil <strong>and</strong> grass that soften the feet, are involvedin producing the disease, along with two bacterial agents, F. necrophorum <strong>and</strong>Bacteroides nodosus. F. necrophorum establishes itself first <strong>and</strong> causes inflammation<strong>and</strong> destruction of the epidermis before penetrating <strong>to</strong> deeper layers. Hoofdegeneration is due <strong>to</strong> the proteolytic properties of B. nodosus. The disease mayappear in several forms: benign, usually caused by less virulent strains of B.nodosus; virulent, with deformation <strong>and</strong> detachment of the hoof; <strong>and</strong> chronic, whichmay last years, with or without producing lameness.Other hoof <strong>diseases</strong> affecting sheep are interdigital dermatitis <strong>and</strong> infectious bulbarnecrosis. The former, caused by F. necrophorum, is characterized by an edema<strong>to</strong>us<strong>and</strong> erythema<strong>to</strong>us inflammation of the interdigital skin, which may be coveredby a layer of moist, gray, necrotic material. Infectious bulbar necrosis is caused byF. necrophorum <strong>and</strong> Corynebacterium pyogenes <strong>and</strong> is characterized by abscesses<strong>and</strong> suppuration of the bulbar area of the hoof, particularly on the hind feet. The diseaseresults from the interaction of both bacteria. C. pyogenes produces a fac<strong>to</strong>r that


192 BACTERIOSESstimulates proliferation of F. necrophorum, <strong>and</strong> the latter protects C. pyogenes fromphagocy<strong>to</strong>sis by producing a leukocidin (Cottral, 1978).CATTLE: Calf diphtheria (necrobacillary s<strong>to</strong>matitis) is characterized by sialorrhea,anorexia, <strong>and</strong> necrotic areas in the oral cavity. Infection can spread <strong>to</strong> the larynx <strong>and</strong>,by inhalation, <strong>to</strong> the lungs, where it causes abscesses <strong>and</strong> pneumonia. The diseaseonly occurs in animals under 2 years of age; mature animals seem immune. The diseaseis caused by F. necrophorum <strong>and</strong> is seen in dairy operations with deficienthygiene. The same disease also affects young goats.Hepatic necrobacillosis is discovered by veterinary inspection in slaughterhouses<strong>and</strong> results in confiscation of carcasses. Lesions on the liver are characterized bywell-delineated yellow areas with a firm consistency.Foot rot in bovines is an acute or chronic necrotic infection of the interdigital skin<strong>and</strong> the coronary region. The chronic form frequently produces arthritis in the distaljoint of the limb. F. necrophorum <strong>and</strong> Bacteroides meleninogenicus have been isolatedfrom biopsy samples of foot rot lesions. A mixture of both bacteria administeredby interdigital scarification or intradermal inoculation reproduced the typicallesions (Berg <strong>and</strong> Loan, 1975). Nevertheless, the etiology still has not been completelyclarified, <strong>and</strong> it is possible that concurrent infection by F. necrophorum <strong>and</strong>other bacteria (B. nodosus, staphylococci) causes the disease (Timoney et al., 1988).Mastitis caused by Bacteroides fragilis has also been described in cattle.SWINE: Pathologies such as ulcerous s<strong>to</strong>matitis, necrotic enteritis, necrotic rhinitis,<strong>and</strong> abscesses have been described in this species.OTHER ANIMAL SPECIES: Similarly <strong>to</strong> what happens in man, osteomyelitis in animalsmay be caused by anaerobes. Of a <strong>to</strong>tal of 39 anaerobic bacteria isolated from19 marrow specimens, the most frequent genus was Bacteroides (18 isolates). B.asaccharolyticus was isolated from 26% of the specimens (Walker et al., 1983).Source of Infection <strong>and</strong> Mode of Transmission: F. necrophorum <strong>and</strong>Bacteroides spp. are part of the normal flora of several mucous membranes inhumans <strong>and</strong> animals. The infection is endogenous, particularly in man. The relativeinfrequency of the disease in man indicates that predisposing fac<strong>to</strong>rs are necessaryfor it <strong>to</strong> occur. These are usually traumas <strong>and</strong> debilitating illnesses. In sum, they areopportunistic agents. A lowered oxidation-reduction potential (E h) resulting frominsufficient blood supply, <strong>to</strong>gether with tissue necrosis <strong>and</strong> the presence of other facultativebacteria, creates a favorable environment for this <strong>and</strong> other anaerobic bacteria.Vascular disease, edema, surgery, <strong>and</strong> cold are some of the <strong>common</strong> fac<strong>to</strong>rsfavoring implantation <strong>and</strong> multiplication of anaerobes (Finegold, 1982). Mostpatients with anaerobic pulmonary infection (abscesses, necrotic pneumonia, pneumonitis,empyema) suffer from altered consciousness or dysphagia due <strong>to</strong> aspirationof the oropharyngeal content, which is rich in anaerobic flora. The underlying conditionsare usually alcoholism, a cerebrovascular accident, general anesthesia, convulsions,<strong>and</strong> narcotics abuse, among others (Bartlett <strong>and</strong> Finegold, 1974).An important predisposing fac<strong>to</strong>r in sheep <strong>and</strong> bovine foot rot is softening of theinterdigital epidermis caused by moist ground, enabling F. necrophorum <strong>to</strong> implantitself <strong>and</strong> multiply. In addition, this bacteria abounds in humid environments (soil<strong>and</strong> grass contaminated by animal feces) <strong>and</strong> has been proved able <strong>to</strong> survive outsidea host’s body for several months. In contrast, B. nodosus is a parasite that can


NECROBACILLOSIS 193live for only a short time in the environment <strong>and</strong> is introduced in establishments bysick or carrier animals. F. necrophorum creates conditions necessary for the multiplicationof B. nodosus. Thus, both bacteria are required <strong>to</strong> cause the disease.As mentioned above, under different conditions other bacteria, such asCorynebacterium pyogenes (which causes infectious bulbar necrosis), interact synergisticallywith F. necrophorum.Bovine hepatic necrobacillosis, the agent of which is F. necrophorum, has anendogenous origin. The agent possibly penetrates by way of the portal circulationfrom epithelial lesions in the rumen, which in turn may be caused by excessive aciditydue <strong>to</strong> provision of concentrated foods.Calf diphtheria, or necrobacillary s<strong>to</strong>matitis, is prevalent in environments wherehygienic practices are markedly poor.Role of Animals in the Epidemiology of the Disease: None. Necrobacillosis isa disease <strong>common</strong> <strong>to</strong> man <strong>and</strong> animals.Diagnosis: When a nonsporulating, anaerobic bacterium is suspected as the causeof infection in a human patient, specimens collected from the lesions for bacteriologicdiagnosis must be free of contaminants from the normal flora, of which theseanaerobes are natural components. Thus, for example, when anaerobic origin is suspectedfor human pulmonary infection, transtracheal aspiration with a needle ordirect penetration of the lung must be used. By contrast, the patient’s sputum is nota suitable material for examination. In the case of empyema or abscesses, obtainingpus under aseptic conditions is not a problem (Finegold, 1982).In veterinary practice, diagnosis of hoof <strong>diseases</strong> in sheep <strong>and</strong> cattle is based onclinical characteristics. Samples for labora<strong>to</strong>ry diagnosis of hepatic necrobacillosiscan be collected without difficulty. In calf diphtheria, ulcerous, necrotizing lesionswith a strong, putrid odor point <strong>to</strong> the disease; if a bacteriologic examination isattempted, epithelial samples from the edges of the ulcer should be used (Guarino etal., 1982).Control: Prevention in man consists primarily of avoiding <strong>and</strong> properly treatingpredisposing conditions. Specific control measures are neither known nor justified.Control of sheep foot rot is the subject of continuing research. An important preventivemeasure is avoiding introduction of animals from places where the diseaseexists, since B. nodosus is considered an obligate parasite. As with other contagious<strong>diseases</strong>, a period of isolation is recommended for recently acquired animals beforeintroducing them in<strong>to</strong> the flock. Once the disease is introduced, transmission maybe reduced by chemoprophylaxis using a foot bath of 5% formalin, 10% zinc sulfate,or 10% copper sulfate. To control the disease, it is recommended that damagedhooves be cut during the dry season in order <strong>to</strong> expose the necrotic parts, <strong>and</strong> thatthe animals be given foot baths or <strong>to</strong>pical treatment with the preparations indicatedabove, in addition <strong>to</strong> intramuscular administration of antibiotics. Studies are stillunder way <strong>to</strong> perfect a vaccine made with B. nodosus, but the existence ofserogroups <strong>and</strong> serotypes within this bacterium complicates this task (Ribeiro,1980). One study indicates that in addition <strong>to</strong> 9 serogroups (A <strong>to</strong> H), there are 16 ormore serotypes. More than one serogroup of B. nodosus may exist in a single flock,<strong>and</strong> several serogroups are sometimes isolated from the hoof of a single sheep.Vaccines made from purified pili (which contain the principal protecting immuno-


194 BACTERIOSESgen) of B. nodosus immunize satisfac<strong>to</strong>rily against a homologous strain of the bacterium(Stewart et al., 1983a). In addition <strong>to</strong> the pili, which only protect againsthomologous strains, there are two other immunogens that could give heterologousimmunity. Vaccines made of whole cells tested against vaccines of purified pili (allhaving an equal pilus content) provided comparable protection. Although vaccinesusing whole cells are cheaper <strong>to</strong> produce <strong>and</strong> confer protection against heterologousstrains, they are irritants <strong>and</strong> cause weight loss (Stewart, 1983b).Control reduces but does not eliminate the problem. Some apparently healthy animalsmay harbor B. nodosus in their hooves <strong>and</strong> maintain the infection in the field.Prevention of calf diphtheria is achieved principally by maintaining hygienicst<strong>and</strong>ards.Chlortetracycline administered in feedlot foods helps <strong>to</strong> reduce the incidence ofhepatic abscesses <strong>and</strong> allows the animals <strong>to</strong> gain weight normally (Timoney et al.,1988). An important preventive measure is not allowing animals <strong>to</strong> change abruptlyfrom their cus<strong>to</strong>mary food <strong>to</strong> concentrated foods.BibliographyBartlett, J.G., S.M. Finegold. Anaerobic infections of the lung <strong>and</strong> pleural space. Am RevResp Dis 110:56–77, 1974.Berg, J.N., R.W. Loan. Fusobacterium necrophorum <strong>and</strong> Bacteroides melaninogenicus asetiologic agents of foot rot in cattle. Am J Vet Res 36:1115–1122, 1975.Clax<strong>to</strong>n, P.D., L.A. Ribeiro, J.R. Eger<strong>to</strong>n. Classification of Bacteroides nodosus by agglutinationtests. Aust Vet J 60:331–334, 1983.Cottral, G.E., ed. Manual of St<strong>and</strong>ardized Methods for Veterinary Microbiology. Ithaca:Coms<strong>to</strong>ck; 1978.Finegold, S.M. Anaerobic bacteria. In: Wyngaarden, J.B., L.H. Smith, Jr., eds. CecilTextbook of Medicine. 16th ed. Philadelphia: Saunders; 1982.Gradin, J.L., J.A. Stephens, G.E. Pluhar, et al. Diversity of pilin of serologically distinctBacteroides nodosus. Am J Vet Res 52:202–205, 1991.Guarino, H., G. Uriarte, J.J. Berreta, J. Maissonnave. Es<strong>to</strong>matitis necrobacilar en terneros.Primera comunicación en Uruguay. In:Primer Congreso Nacional de Veterinaria, Montevideo,Sociedad de Medicina Veterinaria del Uruguay, 1982.Holdeman, L.V., R.W. Kelley, W.E.C. Moore. Anaerobic gram-negative straight, curved <strong>and</strong>helical rods. In: Krieg, N.R., J.G. Holt. Vol I. Bergey’s Systematic Bacteriology. Baltimore:Williams & Wilkins; 1984.Islam, A.K.M.S., J.M. Shneerson. Primary meningitis caused by Bacteroides fragilis <strong>and</strong>Fusobacterium necrophorum. Postgrad Med J 56:351–353, 1980.Jousimies-Somer, H.R., S. Finegold. Anaerobic gram-negative bacilli <strong>and</strong> cocci. In:Ballows, A., W.J. Hausler, Jr., K.L. Hermann, H.D. Isenberg, H.J. Shadomy, eds. Manual ofClinical Microbiology. 5th ed. Washing<strong>to</strong>n, D.C.: American Society for Microbiology; 1991.Kirby, B.D., W.L. George, V.L. Sutter, et al. Gram-negative anaerobic bacilli: Their role ininfection <strong>and</strong> patterns of susceptibility <strong>to</strong> antimicrobial agents. I. Little-known Bacteroidesspecies. Rev Infect Dis 2:914–951, 1980.Ribeiro, L.A.O. Foot-rot dos ovinos: etiología, pa<strong>to</strong>genia e controle. Bol Inst Pesq VetFinamor 7:41–45, 1980.Seidenfeld, S.M., W.L. Sutker, J.P. Luby. Fusobacterium necrophorum septicemia followingoropharyngeal infection. JAMA 248:1348–1350, 1982.Stewart, D.J., B.L. Clark, D.L. Emery, J.E. Peterson, K.J. Fahey. A Bacteroides nodosusimmunogen, distinct from the pilus, which induces cross-protective immunity in sheep vaccinatedagainst footrot. Aust Vet J 60:83–85, 1983a.


NOCARDIOSIS 195Stewart, D.J., B.L. Clark, J.E. Peterson, D.A. Griffiths, E.F. Smith, I.J. O’Donnell. Effec<strong>to</strong>f pilus dose <strong>and</strong> type of Freund’s adjuvant on the antibody <strong>and</strong> protective responses of vaccinatedsheep <strong>to</strong> Bacteroides nodosus. Res Vet Sci 35:130–137, 1983b.Timoney, J.F., J.H. Gillespie, F.W. Scott, J.E. Barlough. Hagan <strong>and</strong> Bruner’s InfectiousDiseases of Domestic Animals. 8th ed. Ithaca: Coms<strong>to</strong>ck; 1988.Walker, R.D., D.C. Richardson, M.J. Bryant, C.S. Draper. Anaerobic bacteria associatedwith osteomyelitis in domestic animals. J Am Vet Med Assoc 182:814–816, 1983.NOCARDIOSISICD-10 A43.0 pulmonary nocardiosis; A43.1 cutaneous nocardiosis;A43.8 other forms of nocardiosisEtiology: Three pathogenic species, Nocardia asteroides, N. brasiliensis, <strong>and</strong> N.otitidiscaviarum (N. caviae). The first was proposed as the type species.Nocardia belong <strong>to</strong> the order Actinomycetales <strong>and</strong> are higher bacteria that resemblefungi in many characteristics. They are aerobic, gram-positive, weakly acid-fast,<strong>and</strong> form long, branched filaments that fragment in<strong>to</strong> coccoid <strong>and</strong> bacillary forms.This fragmentation is the way the bacteria multiply.Geographic Distribution: Worldwide. Nocardia are <strong>common</strong> members of thesoil flora <strong>and</strong> act <strong>to</strong> decompose organic matter. They are not part of the normal floraof man or other animals. There seems <strong>to</strong> be a difference in the distribution of thespecies. N. asteroides has been identified all over the world, while N. brasiliensis ispresent mainly in tropical <strong>and</strong> subtropical climates in North, Central, <strong>and</strong> SouthAmerica (Pier, 1979; L<strong>and</strong> et al., 1991). N. otitidiscaviarum predominates in the soilin the US, India, Japan, Mexico, <strong>and</strong> Tunisia (L<strong>and</strong> et al., 1991).Occurrence in Man: Nocardiosis is not a reportable disease <strong>and</strong> there is no reliableinformation on its frequency. Cases are sporadic. In the US (Beaman et al.,1976), an estimated 500 <strong>to</strong> 1,000 cases occur each year. Between 1972 <strong>and</strong> 1974,81.2% of the cases were due <strong>to</strong> N. asteroides, 5.6% <strong>to</strong> N. brasiliensis, 3% <strong>to</strong> N. otitidiscaviarum,<strong>and</strong> 10.2% <strong>to</strong> unspecified Nocardia. The majority of cases occurredin people between 21 <strong>and</strong> 50 years of age, <strong>and</strong> the male <strong>to</strong> female ratio was 3 <strong>to</strong> 1.Occurrence in Animals: The frequency of animal nocardiosis is not well known.Different <strong>diseases</strong> due <strong>to</strong> Nocardia spp. have been described in cattle, sheep, monkeys,dogs, cats, wild animals, marine mammals, <strong>and</strong> fish. In New Zeal<strong>and</strong>, wherelittle attention had been paid <strong>to</strong> this disease previously, 34 cases were reportedbetween 1976 <strong>and</strong> 1978, <strong>and</strong> 26 of these were manifested as bovine mastitis(Orchard, 1979).The Disease in Man: The principal agent is N. asteroides. Nocardiosis is a suppurativeinfection whose course varies from acute <strong>to</strong> chronic, with a tendency


196 BACTERIOSES<strong>to</strong>ward remission. The most <strong>common</strong> clinical form is pulmonary. Pulmonary nocardiosismay become chronic if not treated properly. Acute pneumonic forms occurprimarily in immunodeficient patients (Lerner, 1991). The symp<strong>to</strong>ma<strong>to</strong>logy is notspecific: cough, respira<strong>to</strong>ry difficulty, <strong>and</strong> hemoptysis when there is chronic cavitation.It usually begins with a primary pyogenous lesion in the lungs. Throughhema<strong>to</strong>genous dissemination, the agent localizes in different organs <strong>and</strong> tissues.Cerebral abscesses are frequent. Between 20% <strong>and</strong> 38% of persons with nocardiosisshow nervous symp<strong>to</strong>ms. The case fatality rate in patients with cerebralabscesses is nearly 50%. A few cases of cerebral abscesses caused by N. otitidiscaviarumhave been reported (Bradsher et al., 1982). Other localizations includesubcutaneous tissue, bones, <strong>and</strong> various organs.Smego <strong>and</strong> Gallis (1984) analyzed 62 cases of infection caused by N. brasiliensisin the US, from their own files <strong>and</strong> from the literature. Of the 62 patients, 46 hadboth a cutaneous disease <strong>and</strong> a soft tissue disease. The cutaneous disease <strong>to</strong>ok theform of cellulitis, pustules, ulcers, pyoderma, subcutaneous abscesses, <strong>and</strong> myce<strong>to</strong>ma.Six patients had a pleuropulmonary disease <strong>and</strong> one of them also had a diseaseof the central nervous system. Dissemination of the disease, which is consideredcharacteristic of N. asteroides, was seen in eight cases. Traumas were animportant predisposing fac<strong>to</strong>r in cutaneous nocardiosis in 19 of 43 cases. All thepatients with cutaneous or soft tissue disease recovered, as did 83% of the pulmonarypatients. Case fatality was high in the cases of dissemination.The recommended treatment is cotrimoxazole, sulfisoxazole, or sulfadiazine. It isimportant that treatment begin as soon as possible <strong>and</strong> continue for some time. Incases that are resistant <strong>to</strong> the sulfonamides, it is advisable <strong>to</strong> add amikacin or highdoses of ampicillin (Benenson, 1990).The incubation period is unknown. It most likely varies depending on the virulence<strong>and</strong> phase of multiplication of the Nocardia strain, as well as the host’s resistance.Most (85%) cases of nocardiosis have occurred in immunologically compromisedpersons (Beaman et al., 1976).N. brasiliensis seldom causes pulmonary disease, but more frequently producesmyce<strong>to</strong>mas.The Disease in Animals: Cattle are the most affected species. N. asteroides <strong>and</strong>,more rarely, N. otitidiscaviarum are agents of bovine mastitis. The udder usuallybecomes infected one <strong>to</strong> two days after calving (Beaman <strong>and</strong> Sugar, 1983), but thedisease may appear throughout lactation, frequently caused by unhygienic therapeuticinfusions in<strong>to</strong> the milk duct. The disease course varies from acute <strong>to</strong> chronic.The mammary gl<strong>and</strong> becomes edema<strong>to</strong>us <strong>and</strong> fibrotic. Fever is <strong>common</strong> <strong>and</strong> prolonged.Pus forms with small granules (microcolonies) as do fistulas <strong>to</strong> the surface.There may also be lymphatic or hema<strong>to</strong>genous dissemination <strong>to</strong> other organs.Among animals with acute infection, mortality is high.Bovine nocardiosis may also manifest as pulmonary disease (especially in calvesunder 6 months of age), abortions, lymphadenitis of various lymph nodes, <strong>and</strong>lesions in different organs.Canids are the second most affected group. The principal agent is N. asteroides,but infections caused by N. brasiliensis <strong>and</strong> N. otitidiscaviarum have also beendescribed. The clinical picture is similar <strong>to</strong> that in man, <strong>and</strong> the most <strong>common</strong> clinicalform is pulmonary. Dogs exhibit fever, anorexia, emaciation, <strong>and</strong> dyspnea.


NOCARDIOSIS 197Dissemination from the lungs <strong>to</strong> other organs is frequent <strong>and</strong> may affect the centralnervous system, bones, <strong>and</strong> kidneys. The cutaneous form is also <strong>common</strong> in dogs,with purulent lesions usually located on the head or extremities. Nocardiosis is mostfrequent in male dogs under 1 year of age. The fatality rate is high (Beaman <strong>and</strong>Sugar, 1983).Nocardiosis in cats is more unusual <strong>and</strong> is seen mostly in castrated males. Mostcases are due <strong>to</strong> N. asteroides, but 30% have been attributed <strong>to</strong> N. brasiliensis or <strong>to</strong>other similar nocardias.A disease with multiple pyogranuloma<strong>to</strong>us foci in the liver, intestines, peri<strong>to</strong>neum,lungs, <strong>and</strong> brain was described in three macaque monkeys (Macacamulatta <strong>and</strong> M. menestrina). Nocardia spp. was isolated in two cases. The assumptionis that two monkeys were infected orally (Liebenberg <strong>and</strong> Giddens, 1985).Earlier references record five cases in monkeys, four of which had a localizedinfection. Pulmonary lesions were found in three of them. N. otitidiscaviarum wasisolated from the h<strong>and</strong> of a baboon (Papio spp.) <strong>and</strong> a cynomolgus macaque (M.fascicularis) had lesions on the brain, jaws, lungs, heart, <strong>and</strong> liver (Liebenberg <strong>and</strong>Giddens, 1985).The recommended treatment is prolonged administration of cotrimoxazole forsome six weeks.Source of Infection <strong>and</strong> Mode of Transmission: Nocardias are componentsof the normal soil flora. These potential pathogens are much more virulent duringthe logarithmic growth phase than during the stationary phase, <strong>and</strong> it is believedthat actively growing soil populations are more virulent for man <strong>and</strong> animals(Orchard, 1979).Man probably acquires the infection by inhaling contaminated dust. Predisposingcauses are important in the pathogenesis of the disease, since most cases occur eitherin persons with deficient immune systems or those taking immunosuppressantdrugs. An outbreak was confirmed among patients in a renal transplant unit <strong>and</strong> thestrain of N. asteroides was isolated from the dust <strong>and</strong> air in the room (Lerner, 1991).Myce<strong>to</strong>mas caused by N. brasiliensis may be caused by a trauma <strong>to</strong> the skin.Wounds that come in<strong>to</strong> contact with the soil may become infected by Nocardia spp.The most <strong>common</strong> route of infection by N. brasiliensis is through traumatic inoculationof the skin by thorns, nails, cat scratches, or burns (Smego <strong>and</strong> Gallis, 1984).Animals probably contract pulmonary infections in the same way as man. Mastitisoccurring later in the lactation period is produced by contaminated catheters.Mastitis at the beginning of lactation is more difficult <strong>to</strong> explain. It is possible thatthe focus of infection already exists in the nonlactating cow <strong>and</strong> that when the udderfills with milk, the infection spreads massively through the milk ducts <strong>and</strong> causesclinical symp<strong>to</strong>ms (Beaman <strong>and</strong> Sugar, 1983). However, the origin of the initialinfection remains an enigma, but it could also be due <strong>to</strong> the insertion of contaminatedinstruments. The multiple cases of nocardia-induced mastitis that are at timesobserved in a dairy herd are attributable <strong>to</strong> transmission of the infection from onecow <strong>to</strong> another by means of contaminated instruments or therapeutic infusions.Role of Animals in the Epidemiology of the Disease: Nocardiosis is a disease<strong>common</strong> <strong>to</strong> man <strong>and</strong> animals; soil is the reservoir <strong>and</strong> source of infection. There areno known cases of transmission from animals <strong>to</strong> man or between humans.


198 BACTERIOSESDiagnosis: Microscopic examination of exudates can indicate nocardiosis, bu<strong>to</strong>nly culture <strong>and</strong> identification of the agent provide a definitive diagnosis. In pulmonarynocardiosis, bronchoalveolar lavage <strong>and</strong> aspiration of abscesses or collectionof fluids can be used, guided by radiology (Forbes et al., 1990).Various serological tests have been described. An enzyme immunoassay with anantigen (a 55-kilodal<strong>to</strong>n protein) specific for Nocardia asteroides yielded goodresults in terms of both sensitivity <strong>and</strong> specificity (Angeles <strong>and</strong> Sugar, 1987).Serodiagnosis in immunodeficient patients—who currently suffer more frequentlyfrom nocardiosis—is very difficult. A more recent work (Boiron <strong>and</strong> Provost, 1990)suggests that the 54-kilodal<strong>to</strong>n protein would be a good c<strong>and</strong>idate as an antigen fora probe for detecting antibodies in nocardiosis.Control: No specific control measures are available. Prevention consists of avoidingpredisposing fac<strong>to</strong>rs <strong>and</strong> exposure <strong>to</strong> dust (Pier, 1979). Environmental hygiene<strong>and</strong> sterilization of instruments are important.For control of mastitis caused by Nocardia spp. in cows, it is recommended thatudder hygiene practices be adopted as well as general hygiene rules for the dairyfacility.BibliographyAngeles, A.M., A.M. Sugar. Rapid diagnosis of nocardiosis with an enzyme immunoassay.J Infect Dis 155:292–296, 1987.Beaman, B.L., J. Burnside, B. Edwards, W. Causey. Nocardial infections in the UnitedStates, 1972–1974. J Infect Dis 134:286–289, 1976.Beaman, B.L., A.M. Sugar. Nocardia in naturally acquired <strong>and</strong> experimental infections inanimals. J Hyg 91:393–419, 1983.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Boiron, P., F. Provost. Use of partially purified 54-kilodal<strong>to</strong>n antigen for diagnosis of nocardiosisby Western blot (immunoblot) assay. J Clin Microbiol 28:328–331, 1990.Bradsher, R.W., T.P. Monson, R.W. Steele. Brain abscess due <strong>to</strong> Nocardia caviae: Repor<strong>to</strong>f a fatal outcome associated with abnormal phagoctye function. Am J Clin Pathol78:124–127, 1982.Forbes, G.M., F.A. Harvey, J.N. Philpott-Howard, et al. Nocardiosis in liver transplantation:Variation in presentation, diagnosis <strong>and</strong> therapy. J Infect 20:11–19, 1990.L<strong>and</strong>, G., M.R. McGinnis, J. Staneck, A. Gatson. Aerobic pathogenic Actinomycetales. In:Balows, A., W.J. Hausler, K.L. Hermann, H.D. Isenberg, H.J. Shadomy, eds. Manual ofClinical Microbiology. 5th ed. Washing<strong>to</strong>n, D.C.: American Society for Microbiology; 1991.Lerner, P.L. Nocardia species. In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E. Bennett, eds.Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: Churchill Livings<strong>to</strong>ne,Inc.; 1990.Liebenberg, S.P., W.E. Giddens. Disseminated nocardiosis in three macaque monkeys. LabAnimal Sci 35:162–166, 1985.Orchard, V.A. Nocardial infections of animals in New Zeal<strong>and</strong>, 1976–78. N Z Vet J27:159–160, 165, 1979.Pier, A.C. Actinomycetes. In: S<strong>to</strong>enner, H., W. Kaplan, M. Torten, eds. Vol 1, Section A:CRC H<strong>and</strong>book Series in Zoonoses. Boca Ra<strong>to</strong>n: CRC Press; 1979.Smego, R.A., H.A. Gallis. The clinical spectrum of Nocardia brasiliensis infection in theUnited States. Rev Infect Dis 6:164–180, 1984.


PASTEURELLOSIS 199PASTEURELLOSISICD-10 A28.0Synonyms: Shipping fever, bovine respira<strong>to</strong>ry disease complex, fibrinous pneumonia(cattle); pasteurella pneumonia (lambs); hemorrhagic septicemia (cattle,lambs); fowl cholera; snuffles (rabbits).Etiology: The genus Pasteurella was reclassified on the basis of DNA:DNAhybridization in order <strong>to</strong> determine the genetic relationship of the different acceptedor proposed species (Mutters et al., 1985). Based on the results of that study, thegenus has been subdivided in<strong>to</strong> 11 species. The species of interest here are:Pasteurella mul<strong>to</strong>cida, P. dagmatis sp. nov., P. canis sp. nov., <strong>and</strong> P. s<strong>to</strong>matis sp.nov. P. caballi, described more recently, should be added as well (Schater et al.,1989). P. haemolytica, an important pathogen for animals <strong>and</strong>, occasionally, forman, is more related <strong>to</strong> the genus Actinobacillus <strong>and</strong> might receive its own genericname in the future (Mutters et al., 1986). In addition, the DNA:DNA hybridizationbetween strains of biotype A <strong>and</strong> biotype T ranges only from 3% <strong>to</strong> 13%, dependingon the biotype used as the reference strain, <strong>and</strong> thus the two biotypes should beclassified as separate species (Bingham et al., 1990). The advantages of reclassificationare not yet evident in epidemiological research, diagnosis, <strong>and</strong> treatment.Pasteurellae are small, pleomorphic, nonmotile, gram-negative, bipolar staining,nonsporulating bacilli, with little resistance <strong>to</strong> physical <strong>and</strong> chemical agents.Subdivision of P. mul<strong>to</strong>cida <strong>and</strong> P. haemolytica in<strong>to</strong> serotypes is important in theareas of epidemiology <strong>and</strong> control (vaccines). Subclassification of P. mul<strong>to</strong>cida in<strong>to</strong>serotypes is based on its capsular (A, B1, D, <strong>and</strong> E) <strong>and</strong> somatic (1–16) antigens;the latter can occur in different combinations. P. haemolytica has been subdividedin<strong>to</strong> two biotypes (A <strong>and</strong> T) <strong>and</strong> 15 serotypes.Geographic Distribution: P. mul<strong>to</strong>cida <strong>and</strong> P. haemolytica are distributedworldwide. The distribution of the other species is less well-known, but based ontheir reservoirs they can be assumed <strong>to</strong> exist on all continents.Occurrence in Man: Rare. It is not a reportable disease <strong>and</strong> its incidence is littleknown. According <strong>to</strong> labora<strong>to</strong>ry records, 822 cases occurred in Great Britain from1956 <strong>to</strong> 1965. A special survey in the US revealed 316 cases caused by P. mul<strong>to</strong>cidafrom 1965 <strong>to</strong> 1968. Data on the occurrence of human pasteurellosis in other countriesare scarce. The disease caused by P. haemolytica is rare.Occurrence in Animals: Common in domestic <strong>and</strong> wild species of mammals<strong>and</strong> birds.The Disease in Man: The principal etiologic agent of human pasteurellosis is P.mul<strong>to</strong>cida. The other species make a lesser contribution <strong>to</strong> human disease. Fifty-sixcultures from Göteborg University (Sweden), obtained from human cases of pasteurellosis,were reexamined. As a result, 26 strains were reclassified as P. mul<strong>to</strong>cidasubspecies mul<strong>to</strong>cida; 11 as P. mul<strong>to</strong>cida ssp. septica; 12 as P. canis; 4 as P.dagmatis, <strong>and</strong> 1 as P. s<strong>to</strong>matis. Two strains were provisionally classified, one as P.haemolytica biogroup 2 (T) <strong>and</strong> another as belonging <strong>to</strong> the group that cannot betyped (Bisgaard <strong>and</strong> Falsen, 1986). The main clinical symp<strong>to</strong>ms of the disease con-


200 BACTERIOSESsist of infected bites or scratches inflicted by cats or dogs (or occasionally by otheranimals), <strong>diseases</strong> of the respira<strong>to</strong>ry system, <strong>and</strong> localized infections in differen<strong>to</strong>rgans <strong>and</strong> tissues. Cases of septicemia are rare. The English-language literaturerecords 21 cases of meningitis (Kumar et al., 1990).Various cases of pasteurellosis in pregnant women have been described. Oneprimigravida who was carrying twins suffered from chorioamnionitis caused by P.mul<strong>to</strong>cida at 27 weeks, after her membranes had broken. The twin close <strong>to</strong> thecervix became infected <strong>and</strong> died shortly after birth, while the other twin did notbecome infected. It is believed that the infection rose upwards from the vagina, withasymp<strong>to</strong>matic colonization (Wong et al., 1992). Two pregnant women with no his<strong>to</strong>ryof concurrent disease received phenoxymethylpenicillin in an early phase ofpasteurellosis. Despite the treatment, one of them became ill with meningitis <strong>and</strong> theother suffered cellulitis with deep abscess formation. Both of them had animals (dog<strong>and</strong> cat), but had not been bitten (Rollof et al., 1992).Most clinical cases arise from infected wounds. Most cats <strong>and</strong> dogs are normalcarriers of Pasteurella <strong>and</strong> harbor the etiologic agent in the oral cavity. The microorganismis transmitted <strong>to</strong> the bite wound <strong>and</strong> a few hours later produces swelling, reddening,<strong>and</strong> intense pain in the region. The inflamma<strong>to</strong>ry process may penetrate in<strong>to</strong>the deep tissue layers, reaching the periosteum <strong>and</strong> producing necrosis. Septicarthritis <strong>and</strong> osteomyelitis are complications that occur with some frequency. Septicarthritis often develops in patients suffering from rheuma<strong>to</strong>id arthritis. Cases havebeen described in which articular complications appeared several months <strong>and</strong> evenyears after the bite (Bjorkholm <strong>and</strong> Eilard, 1983). Of 20 cases of osteomyelitis withor without septic arthritis, 10 developed from cat bites, 5 from dog bites, 1 from dog<strong>and</strong> cat bites, <strong>and</strong> 4 had no known exposure (Ewing et al., 1980).P. mul<strong>to</strong>cida may also aggravate certain respira<strong>to</strong>ry tract <strong>diseases</strong>, such asbronchiectasis, bronchitis, <strong>and</strong> pneumonia. In terms of case numbers, chronic respira<strong>to</strong>ryconditions from which the agent is isolated are second in importance <strong>to</strong> infectiontransmitted by animal bite or scratch. Septicemia <strong>and</strong> endocarditis are extremely rare.The age group most affected is persons over 40 years old, despite the fact thatbites are more frequent in children <strong>and</strong> younger people.P. mul<strong>to</strong>cida is sensitive <strong>to</strong> penicillin, but some resistant animal <strong>and</strong> humanstrains have been found; thus, it is advisable <strong>to</strong> do an antibiogram. In vitro tests havealso shown excellent sensitivity <strong>to</strong> ampicillin, third-generation cephalosporins, <strong>and</strong>tetracycline (Kumar et al., 1990).The Disease in Animals: Pasteurellae have an extremely broad spectrum of animalhosts. Many apparently healthy mammals <strong>and</strong> birds can harbor pasteurellae inthe upper respira<strong>to</strong>ry tract <strong>and</strong> in the mouth. According <strong>to</strong> the most accepted hypothesis,pasteurellosis is a disease of weakened animals that are subjected <strong>to</strong> stress <strong>and</strong>poor hygienic conditions. In an animal with lowered resistance, pasteurellae harboredin the fauces or trachea may become pathogenic for their host. There is amarked difference in the level of virulence among different strains of P. mul<strong>to</strong>cida.In some <strong>diseases</strong>, P. mul<strong>to</strong>cida is the primary <strong>and</strong> only etiologic agent; in others, itis a secondary invader that aggravates the clinical picture.A relationship exists between the serotype of Pasteurella, its animal host, <strong>and</strong> thedisease it causes. Therefore, serologic typing is important for epizootiologic studiesas well as for control (through vaccination).


PASTEURELLOSIS 201Bovine hemorrhagic septicemia is caused by P. mul<strong>to</strong>cida serotype 6:B in Asia,<strong>and</strong> by 6:E <strong>and</strong> 6:B in Africa. In fibrinous pneumonia (“shipping fever”) in cattle,serotype 1 of P. haemolytica, <strong>and</strong> serotype 2:A of P. mul<strong>to</strong>cida predominate.CATTLE: Shipping fever, also called bovine respira<strong>to</strong>ry disease complex, is a syndromethat causes large economic losses in the cattle industry of the WesternHemisphere. In the US, it causes annual losses estimated at more than US$ 25 million.Shipping fever is an acute respira<strong>to</strong>ry disease that particularly affects beefcalves <strong>and</strong> heifers as well as adult cows when they are subjected <strong>to</strong> the stress of prolongedtransport. The symp<strong>to</strong>ma<strong>to</strong>logy varies from a mild respira<strong>to</strong>ry illness <strong>to</strong> arapidly fatal pneumonia. Symp<strong>to</strong>ms generally appear from 5 <strong>to</strong> 14 days after the cattlereach their destination, but some may be sick on arrival. The principal symp<strong>to</strong>msare fever, dyspnea, cough, nasal discharge, depression, <strong>and</strong> appreciable weight loss.The fatality rate is low.The etiology of the disease has not been completely clarified, <strong>and</strong> it is noteworthythat the disease does not occur in Australia, even when animals are transportedover long distances (Irwin et al., 1979). Several concurrent fac<strong>to</strong>rs are believed <strong>to</strong>cause the syndrome. Most prominent among these are such stress fac<strong>to</strong>rs as fatigue,irregular feeding, exposure <strong>to</strong> cold or heat, <strong>and</strong> weaning. Viral infections, whichoccur constantly throughout a herd <strong>and</strong> are often inapparent, are exacerbated by fac<strong>to</strong>rssuch as overcrowding during transport. Moreover, susceptible animals suddenlyadded <strong>to</strong> a herd lead <strong>to</strong> increased virulence. The virus most often identified as theprimary etiologic agent is parainfluenza virus 3 (PI3) of the genus Paramyxovirus.Infection by this virus alone usually causes a mild respira<strong>to</strong>ry disease. However, thedamage it causes <strong>to</strong> the respira<strong>to</strong>ry tract mucosa aids such secondary invaders as P.mul<strong>to</strong>cida <strong>and</strong> P. haemolytica, which aggravate the clinical picture. On the otherh<strong>and</strong>, virulent strains of Pasteurella can cause the disease by themselves.Pasteurellae frequently isolated in cases of shipping fever include P. haemolyticabiotype A, serotype 1, <strong>and</strong> various serotypes of group A of P. mul<strong>to</strong>cida. The factthat treatment with sulfonamides <strong>and</strong> antibiotics gives good results also indicatesthat a large part of the symp<strong>to</strong>ma<strong>to</strong>logy is due <strong>to</strong> pasteurellae. Another importantviral agent that acts synergistically with pasteurellae is the herpesvirus of infectiousbovine rhinotracheitis. Similarly, viral bovine diarrhea, chlamydiae, <strong>and</strong> mycoplasmascan play a part in the etiology of this respira<strong>to</strong>ry disease.An important disease among cattle <strong>and</strong> water buffalo in southern <strong>and</strong> southeasternAsia is hemorrhagic septicemia. In many countries, it is the disease responsiblefor the most losses once rinderpest has been eradicated. Hemorrhagic septicemiaalso occurs in several African countries, including Egypt <strong>and</strong> South Africa, <strong>and</strong>, lessfrequently, in southern Europe. The disease seems <strong>to</strong> be enzootic in American bison,<strong>and</strong> several outbreaks have occurred (the last one in 1967), without the <strong>diseases</strong>preading <strong>to</strong> domestic cattle (Carter, 1982). In tropical countries, hemorrhagic septicemiaoccurs during the rainy season. The main symp<strong>to</strong>ms are fever, edema, sialorrhea,copious nasal secretion, <strong>and</strong> difficulty in breathing. Mortality is high.Surviving animals become carriers <strong>and</strong> perpetuate the disease. Cases of hemorrhagicsepticemia have also been recorded in horses, camels, swine, yaks, <strong>and</strong> otherspecies. It must be borne in mind that hemorrhagic septicemia is due <strong>to</strong> the specificP. mul<strong>to</strong>cida serotypes 6:B <strong>and</strong> 6:E. There is no evidence that the disease occurs indomestic cattle in the Americas.


202 BACTERIOSESP. mul<strong>to</strong>cida is also responsible for cases of mastitis.SHEEP: P. haemolytica is the etiologic agent of two different clinical forms, pneumonia<strong>and</strong> septicemia. Biotype A serotype 2 is the most prevalent agent of pasteurellapneumonia among lambs in Great Britain (Fraser et al., 1982). Pulmonarydisease in sheep follows a viral infection (P13). Although Pasteurella is a secondaryinvader, it is the predominant pathogen. Occurrence of the disease is sporadic orenzootic. The main symp<strong>to</strong>ms are a purulent nasal discharge, cough, diarrhea, <strong>and</strong>general malaise. Lesions consist of hemorrhagic areas in the lungs <strong>and</strong> petechiae inthe pericardium. Pasteurella septicemia is caused by biotype T of P. haemolytica<strong>and</strong> appears in temperate climates in the fall, when the sheep’s diet is changed(Gillespie <strong>and</strong> Timoney, 1981). In Mexico, 860 pneumonic lungs were examined,<strong>and</strong> 120 isolates of P. haemolytica type A were obtained from them. The most <strong>common</strong>serotypes were 1 (22%), 2 (16%), 5 (11%), <strong>and</strong> 9 (7%). Twenty-seven percen<strong>to</strong>f the isolates could not be typed (Colin et al., 1987). P. haemolytica is the only etiologicagent of sporadic sheep mastitis in the western US, Australia, <strong>and</strong> Europe(Blood et al., 1979).SWINE: Pasteurellosis also appears in the form of pneumonia <strong>and</strong>, more rarely, assepticemia. Pasteurella may be a primary or secondary agent of pneumonia, particularlyas a complication of the mild form of classic swine plague (hog cholera) ormycoplasmal pneumonia. The anterior pulmonary lobes are the most affected, withhepatization <strong>and</strong> a sero-fibrinous exudate on the surface. Serotype 3:A of P. mul<strong>to</strong>cidais the most prevalent in chronic swine pneumonia (Pijoan et al., 1983). Studieshave revealed evidence of the etiologic role of <strong>to</strong>xigenic strains of P. mul<strong>to</strong>cidaserotype D in atrophic rhinitis. Bordetella bronchiseptica acting synergistically with<strong>to</strong>xigenic strains of P. mul<strong>to</strong>cida probably causes this disease, the etiology of whichhas been the subject of much debate (Rutter, 1983).Atrophic rhinitis is characterized by atrophy of the nasal turbinate bones, sometimeswith dis<strong>to</strong>rtion of the septum. Experiments have shown that the agents—B. bronchiseptica <strong>and</strong> <strong>to</strong>xigenic P. mul<strong>to</strong>cida—can cause the disease separately in1-week-old gno<strong>to</strong>biotic suckling pigs. However, turbinate atrophy is more severe<strong>and</strong> may become complete when the animals are inoculated with both agents(Rhodes et al., 1987). Atrophic rhinitis could not be seen in some herds from whichonly B. bronchiseptica was isolated. The purified <strong>to</strong>xin of type D strains of P. mul<strong>to</strong>cida,inoculated intranasally, caused severe turbinate atrophy (Dominick <strong>and</strong>Rimler, 1986).Various outbreaks of hemorrhagic septicemia caused by P. mul<strong>to</strong>cida 2:Bhave been reported in India. In one of these outbreaks, 40% of the herd died(Verma, 1988).RABBITS: Pasteurellosis is <strong>common</strong> in rabbit hutches. The most frequent clinicalmanifestation is coryza. As in other animal species, the disease appears under stressfulconditions. The principal symp<strong>to</strong>ms are a serous or purulent exudate from thenose <strong>and</strong> sometimes from the eyes, sneezing, <strong>and</strong> coughing. The pathologicalprocess may spread <strong>to</strong> the lungs. Septicemia <strong>and</strong> death are not un<strong>common</strong>. Malesthat are kept <strong>to</strong>gether may show pasteurella-infected abscesses produced by bites.An atrophic rhinitis syndrome also occurs in rabbits. Au<strong>to</strong>psy of 52 adult rabbitsrevealed that 26 of them (50%) had turbinate atrophy. P. mul<strong>to</strong>cida <strong>and</strong> B. bron-


PASTEURELLOSIS 203chiseptica were isolated from more than 70% of the rabbits. Six percent of thosefrom which only B. bronchiseptica was isolated had the syndrome (DiGiacomo etal., 1989).WILD ANIMALS: Pasteurellosis occurs in many wild animal species, among whichoccasional epizootic outbreaks take place. The etiologic agent is P. mul<strong>to</strong>cida; P.haemolytica has not yet been isolated. Two disease forms are found: hemorrhagicsepticemia, in which the whole animal body is invaded by pasteurellae, <strong>and</strong> the respira<strong>to</strong>rysyndrome or pulmonary pasteurellosis.FOWL: Fowl cholera is an acute septicemic disease with high morbidity <strong>and</strong>morality in all species of domestic fowl. Its incidence has diminished worldwide due<strong>to</strong> improved commercial poultry management practices. The disease usually appearson poultry farms where hygiene is deficient. Explosive outbreaks may occur twodays after infected birds are introduced in<strong>to</strong> a flock. Mortality is variable, at timesreaching 60% of the poultry on a farm. Many of the survivors become carriers <strong>and</strong>give rise <strong>to</strong> new outbreaks. At the beginning of a hyperacute outbreak, fowl die withoutpremoni<strong>to</strong>ry symp<strong>to</strong>ms; mortality increases, but the only symp<strong>to</strong>m seen iscyanosis of the wattle <strong>and</strong> comb. Later, the disease process slows down <strong>and</strong> respira<strong>to</strong>rysymp<strong>to</strong>ms appear. Cases of chronic or localized pasteurellosis may occur followingan acute outbreak, or the disease may take this course from the outset ofinfection. The chronic disease is caused by attenuated strains of P. mul<strong>to</strong>cida <strong>and</strong>manifests itself mostly as “wattle disease” (edematization <strong>and</strong> later caseation ofthese appendages). Another localization can be the wing or foot joints. Fowl cholerais produced by P. mul<strong>to</strong>cida of serogroup A, predominantly serotypes 1 <strong>and</strong> 3(Mushin, 1979); some strains of group D have also been isolated, but they seem <strong>to</strong>be less pathogenic. P. mul<strong>to</strong>cida causes outbreaks with high mortality among wildbirds, especially waterfowl.Source of Infection <strong>and</strong> Mode of Transmission: The reservoir includes cats,dogs, <strong>and</strong> other animals. The etiologic agent is harbored in the upper respira<strong>to</strong>ry passages.Cats are the carriers of the agent 70% <strong>to</strong> 90% of the time, but dogs (20% <strong>to</strong>50%), sheep, cattle, rabbits, <strong>and</strong> rats are also important carriers (Kumar et al., 1990).The most <strong>common</strong> form of the disease (60% <strong>to</strong> 86% of cases) is a wound contaminatedas the result of an animal bite. Cats are primarily responsible in 60% <strong>to</strong> 75%of the cases, followed by dogs. The mode of transmission for the pulmonary form isprobably aerosolization of the saliva of cats or dogs. Some patients (7% <strong>to</strong> 13%) donot acknowledge having been bitten by or otherwise exposed <strong>to</strong> animals (Kumar etal., 1990).For human infections transmitted by animal bite or scratch, the source ofthe infection <strong>and</strong> the mode of transmission are obvious. Except in the case of bites,animal-<strong>to</strong>-man transmission is accomplished through the respira<strong>to</strong>ry or digestivetract. An analysis of 100 cases of human pasteurella infections of the respira<strong>to</strong>rytract <strong>and</strong> other sites found that 69% of the patients had had contact with dogs or cats,or with cattle, fowl, or their products. Nevertheless, 31% of the patients denied allcontact with animals; consequently, it is suspected that interhuman transmissionmay also occur.Among fowl, where P. mul<strong>to</strong>cida is undoubtedly the primary agent of infection,the source of the outbreaks is carrier fowl, <strong>and</strong> transmission occurs predominantly


204 BACTERIOSESby means of aerosols. Dogs <strong>and</strong> cats rarely suffer from pasteurellosis (with theexception of wounds infected with pasteurellae in fights) <strong>and</strong> are healthy carriers.Other mammals acquire the disease from members of their own species eitherthrough the respira<strong>to</strong>ry or digestive tract, or by falling victim <strong>to</strong> the pasteurellae intheir own respira<strong>to</strong>ry tracts when stress lowers their defenses. There is much evidencethat stress fac<strong>to</strong>rs play an important enabling role in unleashing the respira<strong>to</strong>rysyndrome of shipping fever, <strong>and</strong> that these fac<strong>to</strong>rs permit multiplication ofserotype 2 of P. haemolytica (Frank <strong>and</strong> Smith, 1983). Serotypes 6:B <strong>and</strong> 6:E,which cause hemorrhagic septicemia in cattle <strong>and</strong> water buffalo, are perpetuated bymeans of carriers <strong>and</strong> chronically ill animals that serve as a source of infection fortheir kind.Role of Animals in the Epidemiology of the Disease: Pasteurellae survive onlya very short time in the environment. It is certain that animals constitute the mostimportant reservoir of the pasteurellae that are pathogenic for man.Diagnosis: In the case of human infection, diagnosis is made by isolating <strong>and</strong>identifying the etiologic agent from wounds or other sites.In hemorrhagic septicemia or fowl cholera, the etiologic agent can be cultivatedfrom the animal’s blood or viscera. In pneumonia of domestic animals, a pure cultureof pasteurellae may indicate their role in the pathology, but does not revealwhether these bacteria are primary or secondary agents of the disease.Control: Measures <strong>to</strong> reduce the likelihood of bites, such as elimination of straydogs, can prevent some cases of human infection.Control in animals lies mainly in adequate management of herds or poultry farms.Bacterins as well as live attenuated vaccines are in use, or are being tested, againstP. mul<strong>to</strong>cida <strong>and</strong> P. haemolytica. Protection against homologous serotypes is satisfac<strong>to</strong>ry,but protection is only partial or irregular against heterologous serotypes. Ingeneral, attenuated live vaccines give better immunity than bacterins. In Asia, extensiveexperimentation proved that a bacterin with an oil adjuvant can offer solidimmunity against hemorrhagic septicemia. A single dose of live vaccine with astrep<strong>to</strong>mycin-dependent mutant strain conferred immunity against hemorrhagic septicemiain 66.6% <strong>to</strong> 83.3% of calves <strong>and</strong> in 100% of young buffalo (De Alwis <strong>and</strong>Carter, 1980).The use of PI3 vaccine has been recommended for the control of shipping fever.It is better <strong>to</strong> vaccinate against the principal viral agents before weaning or transportinganimals. The bacterins of P. haemolytica <strong>and</strong> P. mul<strong>to</strong>cida have been questioned.Attenuated live vaccines or vaccines from subunits, such as the cy<strong>to</strong><strong>to</strong>xin(leuko<strong>to</strong>xin) of P. haemolytica (Confer et al., 1988), are more reliable. Attenuatedlive vaccines of P. haemolytica are being tested. A bacterin containing multiple antigensof the prevalent serotypes, incorporated in<strong>to</strong> a polyvalent anticlostridial biologicalwith aluminum hydroxide adjuvant, has been tested against P. haemolyticapneumonia in lambs <strong>and</strong> has given satisfac<strong>to</strong>ry results (Wells et al., 1984). Severallive vaccines are available against avian cholera, some of which can be administeredin the drinking water. Selection of Pasteurella strains within the serotypes that causethe disease is important in immunization.Bovine hemorrhagic septicemia should be considered an exotic disease <strong>and</strong>appropriate measures should be taken <strong>to</strong> prevent its spread <strong>to</strong> disease-free areas.


PASTEURELLOSIS 205BibliographyBingham, D.P., R. Moore, A.B. Richards. Comparison of DNA:DNA homology <strong>and</strong> enzymaticactivity between Pasteurella haemolytica <strong>and</strong> related species. Am J Vet Res51:1161–1166, 1990.Bisgaard, M., E. Falsen. Reinvestigation <strong>and</strong> reclassification of a collection of 56 humanisolates of Pasteurellaceae. Acta Pathol Microbiol Immunol Sc<strong>and</strong> [B] 94:215–222, 1986.Bisgaard, M., O. Heltberg, W. Fredriksen. Isolation of Pasteurella caballi from an infectedwound on a veterinary surgeon. Acta Pathol Microbiol Immunol Sc<strong>and</strong> 99(3):291–294, 1991.Bjorkholm, B., T. Eilard. Pasteurella mul<strong>to</strong>cida osteomyelitis caused by cat bite. J Infect6:175–177, 1983.Blood, D.C., J.A. Henderson, O.M. Radostitis. Veterinary Medicine. 5th ed. Philadelphia:Lea <strong>and</strong> Febiger; 1979.Bruner, D.W., J.H. Gillespie. Hagan’s Infectious Diseases of Domestic Animals. 6th ed.Ithaca: Coms<strong>to</strong>ck; 1973.Burdge, D.R., D. Scheifele, D.P. Speert. Serious Pasteurella mul<strong>to</strong>cida infections from lion<strong>and</strong> tiger bites. JAMA 253:3296–3297, 1985.Carter, G.R. Pasteurellosis: Pasteurella mul<strong>to</strong>cida <strong>and</strong> Pasteurella haemolytica [review].Adv Vet Sci 11:321–379, 1967.Carter, G.R. Pasteurella infections as sequelae <strong>to</strong> respira<strong>to</strong>ry viral infections. J Am Vet MedAssoc 163:863–864, 1973.Carter, G.R. Whatever happened <strong>to</strong> hemorrhagic septicemia? J Am Vet Med Assoc180:1176–1177, 1982.Colin, R., L. Jaramillo M., F. Aguilar R., et al. Serotipos de Pasteurella haemolytica en pulmonesneumónicos ovinos en México. Rev Latinoam Microbiol 29:231–234, 1987.Confer, A.W., R.J. Panciera, D.A. Mosier. Bovine pneumonic pasteurellosis: Immunity <strong>to</strong>Pasteurella haemolytica [review]. J Am Vet Med Assoc 193:1308–1316, 1988.De Alwis, M.C., G.R. Carter. Preliminary field trials with a strep<strong>to</strong>mycin-dependent vaccineagainst haemorrhagic septicaemia. Vet Rev 106:435–437, 1980.DiGiacomo, R.F., B.J. Deeb, W.E. Giddens, et al. Atrophic rhinitis in New Zeal<strong>and</strong> whiterabbits infected with Pasteurella mul<strong>to</strong>cida. Am J Vet Res 50:1460–1465, 1989.Dominick, M.A., R.B. Rimler. Turbinate atrophy in gno<strong>to</strong>biotic pigs intranasally inoculatedwith protein <strong>to</strong>xin isolated from type D Pasteurella mul<strong>to</strong>cida. Am J Vet Res 47:1532–1536, 1986.Ewing, R., V. Fainstein, D.M. Musher, M. Lidsky, J. Clarridge. Articular <strong>and</strong> skeletal infectionscaused by Pasteurella mul<strong>to</strong>cida. South Med J 73:1349–1352, 1980.Frank, G.H, R.G. Marshall. Parainfluenza-3 virus infection of cattle. J Am Vet Med Assoc163:858–859, 1973.Frank, G.H., P.C. Smith. Prevalence of Pasteurella haemolytica in transported calves. Am JVet Res 44:981–985, 1983.Fraser, J., N.J. Gilmour, S. Laird, W. Donachie. Prevalence of Pasteurella haemolyticaserotypes isolated from ovine pasteurellosis in Britain. Vet Rec 110:560–561, 1982.Gillespie, J.H., J.F. Timoney. Hagan’s <strong>and</strong> Bruner’s Infectious Diseases of DomesticAnimals. 7th ed. Ithaca: Coms<strong>to</strong>ck; 1981.Harshfield, G.S. Fowl cholera. In: Biester, H.E., L.H. Schwarte, eds. Diseases of Poultry.4th ed. Ames: Iowa State University Press; 1959.Hoerlein, A.B. Shipping fever. In: Gibbons, W.J., ed. Diseases of Cattle. 2nd ed. SantaBarbara: American Veterinary Publications; 1963.Hubbert, W.T., M.N. Rosen. Pasteurella mul<strong>to</strong>cida infection due <strong>to</strong> animal bite. Am JPublic Health 60:1103–1108, 1970.Hubbert, W.T., M.N. Rosen. Pasteurella mul<strong>to</strong>cida infections. II. Pasteurella mul<strong>to</strong>cidainfection in man unrelated <strong>to</strong> animal bite. Am J Public Health 60:1109–1117, 1970.


206 BACTERIOSESIrwin, M.R., S. McConnell, J.D. Coleman, G.E. Wilcox. Bovine respira<strong>to</strong>ry disease complex:A comparison of potential predisposing <strong>and</strong> etiologic fac<strong>to</strong>rs in Australia <strong>and</strong> the UnitedStates. J Am Vet Med Assoc 175:1095–1099, 1979.Kumar, A., H.R. Devlin, H. Vellend. Pasteurella mul<strong>to</strong>cida meningitis in an adult: Casereport <strong>and</strong> review. Rev Infect Dis 12:440–448, 1990.Mair, N.S. Some Pasteurella infections in man. In: Graham-Jones, O., ed. Some Diseasesof Animals Communicable <strong>to</strong> Man in Britain. Oxford: Pergamon Press; 1968.Mushin, R. Serotyping of Pasteurella mul<strong>to</strong>cida isolants from poultry. Avian Dis23:608–615, 1979.Mutters, R., M. Bisgaard, S. Pohl. Taxonomic relationship of selected biogroups ofPasteurella haemolytica as revealed by DNA:DNA hybridizations. Acta Pathol MicrobiolImmunol Sc<strong>and</strong> [B] 94:195–202, 1986.Mutters, R., P. Ihm, S. Pohl, W. Frederiksen, W. Manuheim. Reclassification of the genusPasteurella Trevisan 1887 on the basis of deoxyribonucleic acid homology with proposals forthe new species Pasteurella dagmatis, Pasteurella canis, Pasteurella s<strong>to</strong>matis, Pasteurellaanatis, <strong>and</strong> Pasteurella langaa. Int J Syst Bacteriol 35:309–322, 1985.Namioka, S., M. Murata, R.V.S. Bain. Serological studies on Pasteurella mul<strong>to</strong>cida. V.Some epizootiological findings resulting from O antigenic analysis. Cornell Vet 54:520–534, 1964.Pijoan, C., R.B. Morrison, H.D. Hilley. Serotyping of Pasteurella mul<strong>to</strong>cida isolated fromswine lungs collected at slaughter. J Clin Microbiol 17:1074–1076, 1983.Rhodes, M.B., C.W. New, P.K. Baker, et al. Bordetella bronchiseptica <strong>and</strong> <strong>to</strong>xigenic typeD Pasteurella mul<strong>to</strong>cida as agents of severe atrophic rhinitis of swine. Vet Microbiol13:179–187, 1987.Rollof, J., P.J. Johansson, E. Holst. Severe Pasteurella mul<strong>to</strong>cida infection in pregnantwomen. Sc<strong>and</strong> J Infect Dis 24:453–456, 1992.Rosen, M.N. Pasteurellosis. In: Davis, J.W., L.H. Karstad, D.O. Trainer, eds. InfectiousDiseases of Wild Mammals. Ames: Iowa State University Press; 1970.Rutter, J.M. Virulence of Pasteurella mul<strong>to</strong>cida in atrophic rhinitis of gno<strong>to</strong>biotic pigsinfected with Bordetella bronchiseptica. Res Vet Sci 34:287–295, 1983.Schlater, L.K., D.J. Brenner, A.G. Steigerwalt, et al. Pasteurella caballi,a new species fromequine clinical specimens. J Clin Microbiol 27:2169–2174, 1989.Verma, N.D. Pasteurella mul<strong>to</strong>cida B:2 in haemorrhagic septicaemia outbreak in pigs inIndia. Vet Rec 123:63, 1988.Wells, P.W., J.T. Robinson, N.J. Gilmour, W. Donachie, J.M. Sharp. Development of a combinedclostridial <strong>and</strong> Pasteurella haemolytica vaccine for sheep. Vet Rec 114:266–269, 1984.Wong, G.P., N. Cimolai, J.E. Dimmick, T.R. Martin. Pasteurella mul<strong>to</strong>cida chorioamnionitisfrom vaginal transmission. Acta Obstet Gynecol Sc<strong>and</strong> 71:384–387, 1992.


PLAGUE 207PLAGUEICD-10 A20.0 bubonic plague; A20.2 pneumonic plague;A20.7 septicaemic plagueSynonyms: Black death, pestilential fever, pest.Etiology: The etiologic agent of plague is Yersinia pestis, a gram-negative, nonmotilebacterium, coccobacillary <strong>to</strong> bacillary in form <strong>and</strong> showing bipolar stainingthat is not very resistant <strong>to</strong> physical <strong>and</strong> chemical agents. DNA hybridization studiesdemonstrated the close genetic relationship between Yersinia pestis <strong>and</strong> Y.pseudotuberculosis (Bercovier et al., 1980). On the basis of this, the authors suggestedcalling the etiologic agent of plague Y. pseudotuberculosis subsp. pestis(International Committee on Systemic Bacteriology, List 7, 1981). However, theCommittee’s Judicial Commission (1985) decided <strong>to</strong> reject this nomenclature <strong>and</strong>retain the name Y. pestis in order, among other reasons, <strong>to</strong> avoid possible confusion.Three biological varieties are distinguished: Orientalis (oceanic), Antiqua (continental),<strong>and</strong> Mediaevalis. This distinction has a certain epidemiological significance,principally for nosography, but there is no difference in the biotypes’ pathogenicity.Some virulence fac<strong>to</strong>rs of Y. pestis were defined in the 1980s. Apparently, theprincipal fac<strong>to</strong>r is a 45-megadal<strong>to</strong>n plasmid. This plasmid encodes calcium dependencyfor growth at 37°C, but not at lower temperatures, as well as the virulence antigensV <strong>and</strong> W. The two proteins on the outer membranes that are assumed <strong>to</strong> beimportant in virulence (E <strong>and</strong> K) are also plasmid dependent. The precise role ofeach of these fac<strong>to</strong>rs is not yet well defined (Butler, 1989).Geographic Distribution: Natural foci of infection persist on nearly all continents;they do not exist in Australia, New Zeal<strong>and</strong>, or New Guinea. In the Americas,sylvatic plague is maintained in rodents in the western third of the United States, theborder region of Ecuador <strong>and</strong> Peru, southeastern Bolivia, <strong>and</strong> northeastern Brazil.Similarly, there are foci in north-central, eastern, <strong>and</strong> southern Africa, includingMadagascar; the Near East; the border area between Yemen <strong>and</strong> Saudi Arabia;Kurdistan province (Iran); <strong>and</strong> central <strong>and</strong> Southeast Asia, in Myanmar (Burma) <strong>and</strong>Vietnam. There are also several natural foci in the former Soviet Union <strong>and</strong> inIndonesia (Benenson, 1990).Occurrence in Man: Since the dawn of the Christian era, there have been threegreat p<strong>and</strong>emics: the first began in 542 (Justinian plague) <strong>and</strong> is estimated <strong>to</strong> havecaused 100 million deaths; the second began in 1346, lasted three centuries, <strong>and</strong>claimed 25 million victims; <strong>and</strong> the last began in 1894 <strong>and</strong> continued until the1930s. However, the data on incidence in the Middle Ages are very approximate <strong>and</strong>difficult <strong>to</strong> verify. As a result of the last p<strong>and</strong>emic, natural foci of infection wereestablished in South America, West Africa, South Africa, Madagascar, <strong>and</strong>Indochina.Urban plague has been brought under control in almost the entire world, <strong>and</strong> ruralplague of murine origin is also on the decline. Nevertheless, epidemics haveoccurred in Indonesia, Nepal, <strong>and</strong> southern Vietnam. In this last country, there were5,274 cases in 1967 due <strong>to</strong> contact with domestic rats <strong>and</strong> their fleas.


208 BACTERIOSESFrom 1958 <strong>to</strong> 1979, 46,937 cases of human plague were recorded in 30 countries;if Vietnam is excluded, the <strong>to</strong>tal number is reduced <strong>to</strong> 15,785. The large number ofcases in Vietnam is attributed <strong>to</strong> military operations there <strong>and</strong> consequent ecologicchanges. On the other h<strong>and</strong>, 16 of the 30 countries reporting plague cases were inAfrica. However, incidence of the disease on that continent was very low, less than6% of the world <strong>to</strong>tal (Akiev, 1982). Figure 14 shows the number of cases <strong>and</strong>deaths caused by human plague worldwide from 1971 <strong>to</strong> 1980.The incidence of plague from 1977 <strong>to</strong> 1991 included 14,752 cases with 1,391deaths distributed in 21 countries (<strong>WHO</strong>, 1993).In 1991, there was a large increase of cases in Africa, with a <strong>to</strong>tal of 1,719 peopleaffected, due primarily <strong>to</strong> an outbreak in Tanzania. In that country, there were 60deaths among a <strong>to</strong>tal of 1,293 cases, 1,060 of which occurred in the Tanga region.There were also 137 cases reported in Madagascar <strong>and</strong> 289 in Zaire (<strong>WHO</strong>, 1993).In Asia, there were 226 <strong>to</strong>tal cases, with 15 deaths. There were 100 cases inMyanmar, 94 in Vietnam, 29 in China (with 11 deaths), <strong>and</strong> the remainder in twoother countries (<strong>WHO</strong>, 1993).There are seven countries in the Americas with cases of plague: Bolivia, Brazil,Ecuador, Peru, the US, <strong>and</strong> occasionally, Colombia <strong>and</strong> Venezuela (Akiev, 1982).During the period 1971–1980, there were 2,312 cases in the Americas (Table 2),1,551 of which occurred in Brazil, 316 in Peru, 247 in Bolivia, 123 in the US, <strong>and</strong>75 in Ecuador (<strong>PAHO</strong>, 1981). In all the countries, the number of cases fluctuatedgreatly from year <strong>to</strong> year; at times, epidemic outbreaks have occurred. Plague continues<strong>to</strong> be a public health problem in the Americas because of the persistence ofsylvatic plague <strong>and</strong> the link between domestic <strong>and</strong> wild rodents. In Ecuador, an outbreakof seven cases occurred in May 1976 in Nizac, Chimborazo Province, a settlemen<strong>to</strong>f 850 inhabitants. The outbreak was preceded by a large epizootic in rats<strong>and</strong> mortality among guinea pigs raised in homes for food. The worst outbreak since1966 occurred in 1984 in northern Peru, with 289 cases reported in 40 localities. Anassociation was presumed between this outbreak <strong>and</strong> a great abundance of rodents,possibly the result of ecologic changes due <strong>to</strong> flooding (Rust, 1985).In the US, 35 cases were recorded from April <strong>to</strong> August 1983, the greatest numberof cases since 1925. Almost all the cases occurred in five southwestern states.Twenty-one cases of plague were reported in the Americas in 1991. Ten of theseoccurred in Brazil <strong>and</strong> 11 in the US, although there were no deaths (<strong>WHO</strong>, 1993).In 1992, there were 8 cases in Brazil (all in Bahia) <strong>and</strong> 13 in the US (4 in Arizona,4 in New Mexico, <strong>and</strong> 1 each in five more states) (OPS, 1992). One of the cases inArizona was primary pulmonary plague in a 31-year-old patient who died one dayafter being admitted <strong>to</strong> the hospital. Blood <strong>and</strong> urine cultures taken from the patientwere negative. After the patient’s death, Y. pestis was isolated from the sputum. Thesource of the infection was a sick cat. This is the third case in the US of primary pulmonaryplague contracted from a cat. The incubation period is very short in thesecases (two <strong>to</strong> three days) <strong>and</strong> the symp<strong>to</strong>ms do not lead one <strong>to</strong> suspect plague (CDC,1992). There have been no cases of direct human-<strong>to</strong>-human transmission in the USsince 1924 (Benenson, 1990).In Oc<strong>to</strong>ber 1992, an outbreak of plague was reported in Cajamarca (Peru) whichis still active. In nine localities affected, with an estimated at-risk population of30,000, there were 547 cases <strong>and</strong> 19 deaths (up <strong>to</strong> mid-January 1994). The outbreakswere preceded by deaths among wild rodents <strong>and</strong> guinea pigs (Cavia porcellus) bred


PLAGUE 209Figure 14. Number of cases <strong>and</strong> deaths from human plague worldwide, 1971–1980.2,5002,0004,422 2,755World (cases)World (deaths)Africa (cases)Americas (cases)Asia (cases)1,5001,00050001971 '72 '73 '74 '75 '76 '77 '78 '79 '80Source: <strong>PAHO</strong> Epidemiol Bull 2(6):4–5, 1981.


210 BACTERIOSESTable 2. Number of cases <strong>and</strong> deaths from human plague in the Americas, 1971–1980.Country ________ 1971 ________ 1972 ________ 1973 ________ 1974 ________ 1975 ________ 1976 ________ 1977 ________ 1978 ________ 1979 ________ 1980C D C D C D C D C D C D C D C D C D C DBolivia 19 3 0 0 0 0 14 5 2 0 24 5 29 9 68 2 10 0 26 2Brazil 146 2 169 13 152 ... 291 ... 496 5 97 ... 1 ... 11 ... 0 0 98 0Ecuador 27 0 9 0 1 1 0 0 0 0 8 1 0 0 0 0 0 0 0 0Peru 22 5 118 15 30 2 8 2 3 0 1 0 0 0 6 1 0 0 0 0United Statesof America a 2 0 1 0 2 0 8 1 20 4 16 3 18 2 12 2 13 2 18 5Total 216 10 297 28 185 3 321 8 521 9 146 9 48 11 97 5 23 2 142 7C = CasesD = Deaths… Data unavailablea Plague found in rodents.Source: <strong>PAHO</strong> Epidemiol Bull 2(6):4–5, 1981.


PLAGUE 211at home by the peasants. One fac<strong>to</strong>r that helped <strong>to</strong> increase the number of cases wasthat rodenticides were used without simultaneous or prior use of flea pulicides(Report from Dr. Alfonso Ruiz <strong>to</strong> the Pan American Health Organization, February8, 1994).Occurrence in Animals: Natural infection by Y. pestis has been found in 230species <strong>and</strong> subspecies of wild rodents. In natural foci, sylvatic plague is perpetuatedthrough the continuous circulation of the etiologic agent, transmitted by fleasfrom one rodent <strong>to</strong> another. It is generally believed that the survival of the etiologicagent in a natural focus depends on the existence of rodent species, or individualswithin a species, with differing levels of susceptibility. The most resistant individualsare host <strong>to</strong> <strong>and</strong> infect the fleas, which in turn infect susceptible animals in thearea <strong>and</strong> can spread <strong>to</strong> domestic rodents. Susceptible animals generally die, but theyincrease the population of infected fleas by means of their bacteremia. When thenumber of susceptible individuals is large <strong>and</strong> climatic conditions favorable, an epizooticmay develop in which many rodents die. As the epizootic diminishes, theinfection continues in enzootic form in the surviving population until a new outbreakoccurs. Infection may remain latent in enzootic foci for a long time, <strong>and</strong> theabsence of human cases should not be interpreted as a sign that the natural focus iseliminated.During the period 1966–1982, 861 isolates were taken of Y. pestis in foci in northeasternBrazil. Of these, 471 were from rodents or other small mammals, 236 werefrom batches of fleas, 2 from batches of Ornithodorus, <strong>and</strong> 152 from patients. In therodents, the highest number of isolates were taken from Zygodon<strong>to</strong>mys lasiarus pixuna,which also provided the highest number of fleas, primarily of the genusPolygenis; on only one occasion was the agent isolated from cat fleas(Ctenocephalides felis). The agent was isolated from human fleas (Pulex irritans)found on the floor of dwellings on 10 occasions. Human flea infection suggests thepossibility of human-<strong>to</strong>-human transmission through flea bites, usually after a fatalcase in the family (Almeida et al., 1985).House cats that come in<strong>to</strong> contact with rodents <strong>and</strong>/or their fleas can becomeinfected <strong>and</strong> fall ill, <strong>and</strong> can transmit the infection <strong>to</strong> man. In the US <strong>and</strong> SouthAfrica, several cases of the disease in cats have been described (Kaufmann et al.,1981; Rollag et al., 1981). In New Mexico (USA), 119 cases of plague werereported in domestic cats from 1977 <strong>to</strong> 1988 (Eidson et al., 1991). There is also evidencethat camels <strong>and</strong> sheep in enzootic plague areas can contract the infection <strong>and</strong>that, in turn, man can become infected when sacrificing these animals. Such casesoccurred in Libya (Christie et al., 1980).The Disease in Man: The incubation period lasts from two <strong>to</strong> six days, though itmay be shorter. Three clinical forms of plague are recognized: bubonic, septicemic,<strong>and</strong> pneumonic. The symp<strong>to</strong>ms shared by all three are fever, chills, cephalalgia, nausea,generalized pain, diarrhea, or constipation; <strong>to</strong>xemia, shock, arterial hypotension,rapid pulse, anxiety, staggering gait, slurred speech, mental confusion, <strong>and</strong>prostration are also frequent.Bubonic plague—the most <strong>common</strong> form in interp<strong>and</strong>emic periods—is characterizedby acute inflammation <strong>and</strong> swelling of peripheral lymph nodes (buboes),which can become suppurative. There may be a small vesicle at the site of the fleabite. The buboes are painful <strong>and</strong> the surrounding area is usually edema<strong>to</strong>us.


212 BACTERIOSESBacteremia is present at the beginning of the disease. The fatality rate in untreatedcases is from 25% <strong>to</strong> 60%. At times, the disease may take the form of a mild, localized,<strong>and</strong> short-lived infection (pestis minor). Another, less frequent form is meningitis,which occurs primarily after ineffective treatment for bubonic plague (Butler,1988). In septicemic plague, nervous <strong>and</strong> cerebral symp<strong>to</strong>ms develop extremely rapidly.Epistaxis, cutaneous petechiae, hematuria, <strong>and</strong> involuntary bowel movementsare seen. The course of the disease is very rapid, from one <strong>to</strong> three days, <strong>and</strong> casefatality may reach nearly 100%.Pneumonic plague may be a secondary form derived from the bubonic or septicemicforms by hema<strong>to</strong>genous dissemination, or it may be primary, produceddirectly by inhalation during contact with a pneumonic plague patient (primarypneumonic plague). In addition <strong>to</strong> the symp<strong>to</strong>ms <strong>common</strong> <strong>to</strong> all forms, dyspnea,cough, <strong>and</strong> expec<strong>to</strong>ration are present. The sputum may vary from watery <strong>and</strong> foamy<strong>to</strong> patently hemorrhagic. This is the most serious form.Primary pneumonic plague, the origin of which is human-<strong>to</strong>-human transmissionby aerosol <strong>and</strong> which has caused outbreaks <strong>and</strong> sometimes devastating epidemics,is rare. The pneumonic form seen in present times is the secondary form, resultingfrom septicemic dissemination. Since 1925, the US has recorded very few casesof primary pneumonic plague, all of which have resulted from exposure <strong>to</strong> a catwith secondary pneumonia. The first case occurred in California in 1980 (CDC,1982). A similar case occurred more recently in Arizona (CDC, 1992). In <strong>to</strong>tal,there have been three cases of primary pneumonia with the same characteristics.Secondary invasion of the lungs (secondary pneumonic plague) occurs in untreatedpatients <strong>and</strong> approximately 95% of them die without becoming transmitters ofthe agent by aerosol. If left untreated, the small number of patients who do not diemay give rise <strong>to</strong> other cases of pneumonic plague by airborne transmission (Pol<strong>and</strong><strong>and</strong> Barnes, 1979). In countries that maintain epidemiologic surveillance <strong>and</strong>where physicians <strong>and</strong> the general population are alert <strong>to</strong> the disease, the high fatalityrates caused by all forms of plague have been largely arrested by early diagnosis<strong>and</strong> prompt treatment with antibiotics, such as strep<strong>to</strong>mycin, tetracycline, <strong>and</strong>chloramphenicol.The Disease in Animals: Y. pestis primarily infects animals of the orderRodentia; if affects wild as well as domestic rodents <strong>and</strong>, <strong>to</strong> a lesser degree, rabbits<strong>and</strong> hares (lagomorphs). The infection may be acute, chronic, or inapparent.Different species of rodents <strong>and</strong> different populations of the same species showvarying degrees of susceptibility. In this regard, it has been observed that a populationin an enzootic area is more resistant than another in a plague-free area, a phenomenonattributed <strong>to</strong> natural selection. Domestic (commensal) rats are very susceptible;Rattus rattus die in large numbers during epizootics. By contrast,susceptibility varies greatly between different species in natural foci <strong>and</strong> must bedetermined for each situation. In the western United States, prairie dogs (Cynomysspp.) <strong>and</strong> the ground squirrel Citellus beecheyi are very susceptible, while certainspecies of Microtus or Peromyscus are resistant.Lesions found in susceptible animals dead from plague vary with the course of thedisease. In acute cases, hemorrhagic buboes <strong>and</strong> splenomegaly are present withou<strong>to</strong>ther internal lesions; in subacute cases the buboes are caseous, <strong>and</strong> punctiformnecrotic foci are found in the spleen, liver, <strong>and</strong> lungs.


PLAGUE 213Natural infection in cats has come under close scrutiny, as they have been a sourceof infection for man in several instances. Feline plague is characterized by formationof abscesses, lymphadenitis, lethargy, <strong>and</strong> fever (Rollag et al., 1981). Secondary pneumoniamay also be present, as in the case at Lake Tahoe, California, where a kittentransmitted the infection <strong>to</strong> a man by aerosol. Fatality is over 50% in cats infectedexperimentally. In contrast, dogs inoculated with the plague agent react only withfever. Other carnivores are not very susceptible, with the exception of individuals withgreater than normal susceptibility, as might be expected in any animal population.Natural infection has been recorded in camels <strong>and</strong> sheep in the former SovietUnion <strong>and</strong> Libya (Christie et al., 1980) <strong>and</strong>, more recently, in camels from SaudiArabia (A. Barnes, personal communication).Source of Infection <strong>and</strong> Mode of Transmission (Figure 15): Wild rodents arethe natural reservoir. The maintenance hosts vary in each natural focus, but they arealmost always rodent species with low susceptibility, i.e., the animals becomeinfected but do not die from the disease. Very susceptible species, in which manyanimals die during an epizootic, are important in amplification <strong>and</strong> diffusion of theinfection as well as in its transmission <strong>to</strong> man, but they cannot be permanent hosts.The epizootics that afflict prairie dogs (Cynomys spp.) are devastating. In one epizootic,only some animals survived in two of seven colonies. Another explosive epizooticannihilated an entire colony of 1,000 <strong>to</strong> 1,500 animals in two months. A thirdepizootic reduced the population by 85% (Ubico et al., 1988). R. rattus is very susceptible,but the infection usually dies out rapidly in this species. Only in some circumstances,as occurred in India, can it serve as a temporary host, but not for manyyears. Consequently, the persistence of a focus depends on rodent species that havea wide spectrum of partial resistance.In a natural focus, the infection is transmitted from one individual <strong>to</strong> another byfleas. Different species of fleas vary greatly in their efficiency as vec<strong>to</strong>rs. BiologicalFigure 15. Plague. Domestic <strong>and</strong> peridomestic transmission cycle.InfectedrodentsTick biteXenopsylla cheopis(rat flea)BiteSusceptiblerodentsBiteMan


214 BACTERIOSESvec<strong>to</strong>rs are characterized by the blocking phenomenon. When Y. pestis is ingestedwith the septicemic host’s blood, the agent multiplies in the flea’s s<strong>to</strong>mach <strong>and</strong> theproventriculum becomes blocked by the mass of bacteria. When a blocked flea tries <strong>to</strong>feed again, it regurgitates the bacteria in<strong>to</strong> the bloodstream of the new host (this is thecase with Xenopsylla cheopis, the domestic rat flea). Wild rodent fleas are generallyless efficient <strong>and</strong> their capacity as biological vec<strong>to</strong>rs varies; it is believed that mechanicaltransmission may be important in natural foci. Also, these vec<strong>to</strong>rs are not veryspecies-specific <strong>and</strong> can transmit the infection between different rodent species livingin an enzootic area. The etiologic agent survives for a long time in fleas; some haveremained infected for a period of 396 days. For this reason, fleas may be consideredpart of the natural reservoir, which would be an arthropod-vertebrate complex. Morethan 200 species of fleas have been implicated in the transmission of plague.Infection from a natural focus may be passed <strong>to</strong> commensal rodents (domestic rats<strong>and</strong> mice) by members of the ubiqui<strong>to</strong>us rodent species that approach hum<strong>and</strong>wellings <strong>and</strong> can thus initiate an outbreak of plague within households. In the sameway, peridomestic rodents may come in<strong>to</strong> contact with wild rodents. Transmissionis effected by means of fleas.Other mammals (dogs, marsupials) may also serve as the link between the wild<strong>and</strong> domestic cycles by transporting fleas from one place <strong>to</strong> another. In northeasternBrazil, South American short-tailed gray opossums (Monodelphis domestica) naturallyinfected with the plague agent via Polygenis bohlsi jordani (a principal vec<strong>to</strong>rof sylvatic plague in this region) have been found <strong>to</strong> live near <strong>and</strong> enter houses. Thenatural plague foci can experience long periods of reduced activity, during which theproportion of infected rodents is small <strong>and</strong> no human cases occur. When these focibecome active, epizootics among rodents <strong>and</strong>, at times, epidemic outbreaks canoccur. Such could have been the case in the central Java (Indonesia) focus where nohuman plague had occurred since 1959, but where 100 cases were reported in 1968<strong>and</strong> 40 in 1971.When man enters a natural focus, he may contract the infection through bites offleas of wild rodents or lagomorphs, or through skin abrasions or bites when h<strong>and</strong>lingthese animals. Human cases are sporadic under these circumstances. Whenplague penetrates the domestic <strong>and</strong> peridomestic environment, man is infected viafleas of commensal rodents, <strong>and</strong> epidemic outbreaks may result. The domestic ratflea (Xenopsylla cheopsis) is the biological vec<strong>to</strong>r par excellence of plague. Thename zootic plague has been given <strong>to</strong> plague transmitted by insects. Indirect interhumantransmission via human ec<strong>to</strong>parasites (Pulex irritans <strong>and</strong> Pediculus humanis)is rare <strong>and</strong> has only been observed in heavily infected environments. In some areasof the Andes, this mode of transmission occurs with some frequency, especially duringwakes for those who have died of plague. These outbreaks almost always occurwithin families.Secondary pneumonia as a complication of bubonic or septicemic plague maygive rise <strong>to</strong> a series of primary pneumonic plague cases through interhuman transmissionvia the respira<strong>to</strong>ry route. This is so-called demic plague. At present, bubonicplague is eminently zoonotic <strong>and</strong> occurs primarily in semi-arid areas.Cats have transmitted the infection in a small proportion of cases (in the US, 2.2%from 1930 <strong>to</strong> 1979). Because buboes in cats are located in the head <strong>and</strong> neck region,it is thought that cats contract the infection by consuming infected rodents.Transmission from cat <strong>to</strong> man has resulted from direct contact, bites, or scratches.


PLAGUE 215Role of Animals in the Epidemiology of the Disease: Perpetuation of plaguedepends on the Y. pestis-rodents-fleas complex in natural foci. Plague in commensalrats is usually a collateral phenomenon <strong>to</strong> sylvatic plague, <strong>and</strong> so, by extension,is demic plague.Diagnosis: Early diagnosis is essential <strong>to</strong> protect the patient <strong>and</strong> the community.Diagnosis is confirmed in the labora<strong>to</strong>ry by puncturing the bubo <strong>and</strong> collecting fluidfrom gelatinous edemas, cerebrospinal fluid, <strong>and</strong> sputum for preparation of a GramorGiemsa-stained smear, <strong>and</strong> culturing in appropriate media. The culture can beidentified rapidly using specific phagocy<strong>to</strong>lysis or the immunofluorescence <strong>and</strong>agglutination tests.An index case (the first case in a community), which may be the precursor of anoutbreak, can be provisionally diagnosed with the rapid immunofluorescence test,using material from a bubo, <strong>and</strong> confirmed later by culture or inoculation in labora<strong>to</strong>ryanimals (guinea pigs or mice).Hemoculture can be used in the initial, septicemic period of bubonic plague.The serological tests most often used for human patients are passive hemagglutination<strong>and</strong> the fluorescent antibody test. The enzyme-linked immunosorbent assay(ELISA) procedure for detecting the F1 antigen (Fraction 1) of Y. pestis with monoclonalantibodies yields apparently satisfac<strong>to</strong>ry results, but does not eliminate theneed for bacteriological confirmation (Williams et al., 1986).Inoculation of labora<strong>to</strong>ry animals has proven superior <strong>to</strong> culture on culture mediafor plague research in rodents or fleas. Passive hemagglutination is of great value forepizootic studies of the infection, both in native rodent populations <strong>and</strong> in sentinelanimals in natural foci. Resistant animals, such as dogs, can fulfill the latter surveillancefunction. During a plague episode in which one man in southeastern Utah(USA) died, the only evidence of the infection’s activity was the discovery of positivetiters in two of the family dogs. In the same country, coyotes have proved usefulas sentinels. Coyotes rarely die of plague, but produce antibodies against the diseaseagent; in addition, since they feed on sick <strong>and</strong> dead rodents, examining a coyoteis equivalent <strong>to</strong> examining several hundred rodents. A rapid serological test (anenzymatic immunoassay) has been perfected for testing these animals (Willeberg etal., 1979). The passive hemagglutination test, employing specific Fraction-1 antigen(pesticin), is also useful in retrospective studies of plague in human communities inenzootic areas. A DNA probe has been developed that could prove useful in epidemiologicalsurveillance of plague (McDonough et al., 1988).Control: Prevention of human plague is based on control of rodents <strong>and</strong> vec<strong>to</strong>rsof infection. Eradication of natural foci is a long-term, costly, <strong>and</strong> difficult task thatcan be achieved by changing the ecology of the foci <strong>and</strong> dedicating the enzootic area<strong>to</strong> agriculture. In general, the objectives of prevention campaigns are more limited<strong>and</strong> consist mainly of emergency programs in situations with a high potential forhuman infection. In all areas where natural plague foci exist, continuous surveillancemust be maintained (dogs have been used very successfully as sentinel animals)<strong>and</strong> emergency measures set in motion if cases of the disease develop.Essentially, these measures consist of the use of insecticides <strong>and</strong> rodenticides.Insecticides should be employed before or at the same time as rodenticides, butnever after, as fleas ab<strong>and</strong>on dead animal hosts <strong>and</strong> seek out new hosts, includingman. During outbreaks, the main effort should be directed <strong>to</strong>ward flea control,


216 BACTERIOSESwhich is very effective <strong>and</strong> economical. If human plague cases occur, patients mustbe isolated (stringent isolation is required for pneumonic patients) <strong>and</strong> treated. Allcontacts should be disinfected <strong>and</strong> kept under surveillance; if deemed necessary,chemoprophylaxis (tetracycline <strong>and</strong> sulfonamides) should be given for six days; flea<strong>and</strong> rodent control should be continued. In such places as the Andes, where fleainfestations on humans are prevalent, prophylactic measures are recommended forpersons attending funerals of plague victims, along with strict control of these cases<strong>to</strong> prevent human-<strong>to</strong>-human transmission.In the mountains of Tienshan (China), measures were taken <strong>to</strong> control the gray orAltai marmot (Marmota baibacina), a reservoir of plague. Between 1967 <strong>and</strong> 1987,the marmot population was reduced from 14.52 animals for every 10 hectares in1967 <strong>to</strong> 0.91 in 1987. More recently, bacteriologic <strong>and</strong> serologic tests were performedon 5,000 marmots <strong>and</strong> 2,000 domestic dogs; with the exception of threedogs, the tests were negative. No more human cases were reported (Lu et al., 1991).The inactivated vaccine provides protection for no more than six months <strong>and</strong> vaccinationis justified only for inhabitants of high-incidence areas, labora<strong>to</strong>ry personnelwho work with plague, <strong>and</strong> people who must enter a plague focus. It should bekept in mind that several doses are needed <strong>to</strong> obtain a satisfac<strong>to</strong>ry level of protection.The inactivated vaccine was used on US troops in Vietnam <strong>and</strong> is believed <strong>to</strong>have been very useful in protecting them.Plague is subject <strong>to</strong> control measures established under the International SanitaryCode (World Health Organization).BibliographyAkiev, A.K. Epidemiology <strong>and</strong> incidence of plague in the world, 1958–79. Bull WorldHealth Organ 60:165–169, 1982.Almeida, A.M.P. de., D.P. Brasil, F.G. de Carvalho, C.R. de Almeida. Isolamen<strong>to</strong> daYersinia pestis nos focos pes<strong>to</strong>sos do nordeste do Brasil no periodo de 1966 a 1982. Rev InstMed Trop Sao Paulo 27:207–218, 1985.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bercovier, H., H.H. Mollaret, J.M. Alonso, J. Brault, G.R. Fanning, A.G. Steigerwalt, et al.Intra- <strong>and</strong> interspecies relatedness of Yersinia pestis by DNA hybridization <strong>and</strong> its relationship<strong>to</strong> Yersinia pseudotuberculosis. Curr Microbiol 4:225–229, 1980.Butler, T. Plague. In: Warren, K.S., A.A.F. Mahmoud, eds. Tropical <strong>and</strong> GeographicalMedicine. New York: McGraw-Hill; 1984.Butler, T. The black death past <strong>and</strong> present. 1. Plague in the 1980s. Trans R Soc Trop MedHyg 83:458–460, 1989.Christie, A.B., T.H. Chen, S.S. Elberg. Plague in camels <strong>and</strong> goats: Their role in human epidemics.J Infect Dis 141:724–726, 1980.Davis, D.H.S., A.F. Hallett, M. Isaacson. Plague. In: Hubbert, W.T., W.F. McCulloch, P.R.Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield:Thomas; 1975.Dinger, J.E. Plague. In:Van der Hoeden, J., ed. Zoonoses. Amsterdam: Elsevier; 1964.Eidson, M., J.P. Thilsted, O.J. Rollag. Clinical, clinicopathologic, <strong>and</strong> pathologic featuresof plague in cats: 119 cases (1977–1988). J Am Vet Med Assoc 199:1191–1197, 1991.Hudson, B.W., M.I. Goldenberg, J.D. McCluskie, H.E. Larson, C.D. McGuire, A.M.


PLAGUE 217Barnes, et al. Serological <strong>and</strong> bacteriological investigations of an outbreak of plague in anurban tree squirrel population. Am J Trop Med Hyg 20:225–263, 1971.International Committee on Systemic Bacteriology, List 7. Validation of the publication ofnew names <strong>and</strong> new combinations previously effectively published outside USB. Int J SystBacteriol 31:382–383, 1981.Judicial Commission of the International Commitee on Systemic Bacteriology. Opinion 60.Rejection of the name Yersinia pseudotuberculosis subsp. Yersinia pestis (Lehmann <strong>and</strong>Neumann) van Loghem 1944 for the plague bacillus. Int J Syst Bacteriol 35:540, 1985.Kartman, L., M.I. Goldenberg, W.T. Hubbert. Recent observations on the epidemiology ofplague in the United States. Am J Public Health 56:1554–1569, 1966.Kaufmann, A.F., J.M. Mann, T.M. Gardiner, F. Hea<strong>to</strong>n, J.D. Pol<strong>and</strong>, A.M. Barnes, et al.Public health implications of plague in domestic cats. J Am Vet Med Assoc 179:875–878, 1981.Lu, C.F. [Epidemiologic significance of the eradication of the gray marmot (Marmotabaibacina) in natural foci in the mountains of Tienshan, in Hutubi District, Xinjang]. BullEndem Dis 5:4–18, 1990–1991.McDonough, K.A., T.G. Schwan, R.E. Thomas, S. Falkow. Identification of a Yersiniapestis-specific DNA probe with potential for use in plague surveillance. J Clin Microbiol26:2515–2519, 1988.Meyer, K.F. Pasteurella <strong>and</strong> Francisella. In: Dubos, R.J., J.G. Hirsch, eds. Bacterial <strong>and</strong>Mycotic Infections of Man. 4th ed. Philadelphia: Lippincott; 1965.Olsen, P.F. Sylvatic (wild rodent) plague. In: Davis, J.W., L.H. Karstad, D.O. Trainer, eds.Infectious Diseases of Wild Mammals. Ames: Iowa State University Press; 1970.Organización Panamericana de la Salud (OPS). Enfermedades sujetas al Reglamen<strong>to</strong>Sanitario Internacional. Bol Epidemiol 13:16, 1992.Pan American Health Organization (<strong>PAHO</strong>). Health Conditions in the Americas,1969–1972. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1974. (Scientific Publication 287).Pan American Health Organization (<strong>PAHO</strong>). Status of plague in the Americas, 1970–1980.Epidemiol Bull 2:5–8, 1981.Pan American Health Organization (<strong>PAHO</strong>). Plague in the Americas. Washing<strong>to</strong>n, D.C.:<strong>PAHO</strong>; 1965. (Scientific Publication 115).Pavlovsky, E.N. Natural Nidality of Transmissible Diseases. Urbana: University of IllinoisPress; 1966.Pol<strong>and</strong>, J.D., A.M. Barnes. Plague. In: S<strong>to</strong>enner, H., W. Kaplan, M. Torten, eds. Vol 1,Section A: CRC H<strong>and</strong>book Series in Zoonoses. Boca Ra<strong>to</strong>n: CRC Press; 1979.Pollitzer, R. A review of recent literature on plague. Bull World Health Organ 23:313–400, 1960.Pollitzer, R., K.F. Meyer. The ecology of plague. In: May, J.M., ed. Studies in DiseaseEcology. New York: Hafner Pub. Co.; 1961.Rollag, O.J., M.R. Skeels, L.J. Nims, J.P. Thilsted, J.M. Mann. Feline plague in NewMexico: Report of five cases. J Am Vet Med Assoc 179:1381–1383, 1981.Rust, J.H. Plague research in northern Peru. <strong>PAHO</strong>/<strong>WHO</strong> report, June 1985.Stark, H.E., B.W. Hudson, B. Pittman. Plague Epidemiology. Atlanta: US Centers forDisease Control <strong>and</strong> Prevention; 1966.Tirador, D.F., B.E. Miller, J.W. Stacy, A.R. Martin, L. Kartman, R.N. Collins, et al. Plagueepidemic in New Mexico, 1965. An emergency program <strong>to</strong> control plague. Public Health Rep82:1094–1099, 1967.Ubico, S.R., G.O. Maupin, K.A. Fagers<strong>to</strong>ne, R.G. McLean. A plague epizootic in the whitetailedprairie dogs (Cynomys leucurus) of Meeteetse, Wyoming. J Wildl Dis 24:399–406, 1988.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Human plague—United States, 1981. MMWR Morb MortalWkly Rep 31:74–76, 1982.


218 BACTERIOSESUnited States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Pneumonic plague—Arizona, 1992. MMWR Morb MortalWkly Rep 41:737–739, 1992.Willeberg, P.W., R. Ruppanner, D.E. Behymer, H.H. Higa, C.E. Franti, R.A. Thomson, etal. Epidemiologic survey of sylvatic plague by serotesting coyote sentinels with enzymeimmunoassay. Am J Epidemiol 110:328–334, 1979.Williams, J.E., L. Arntzen, G.L. Tyndal, M. Isaacson. Application of enzyme immunoassaysfor the confirmation of clinically suspect plague in Namibia, 1982. Bull World HealthOrgan 64:745–752, 1986.World Health Organization (<strong>WHO</strong>). <strong>WHO</strong> Expert Committee on Plague. Fourth Report.Geneva: <strong>WHO</strong>; 1970. (Technical Report Series 447).World Health Organization (<strong>WHO</strong>). Human plague in 1991. Wkly Epidemiol Rec68(4):21–23, 1993.PSEUDOTUBERCULOUS YERSINIOSISICD-10 A28.2 extraintestinal yersiniosisEtiology: Yersinia pseudotuberculosis is a coccobacillary, gram-negative bacteriathat is motile at 25°C, nonmotile at 37°C, <strong>and</strong> can live a long time in soil <strong>and</strong> water.It belongs <strong>to</strong> the family Enterobacteriaceae. DNA hybridization studies have confirmedthe close relationship between the agent of plague <strong>and</strong> that of pseudotuberculousyersiniosis.Y. pseudotuberculosis is subdivided on the basis of its biochemical properties in<strong>to</strong>five biotypes <strong>and</strong> on the basis of somatic (O) antigens in<strong>to</strong> six serogroups (1–6),types 1, 2, 4, <strong>and</strong> 5 of which are divided in<strong>to</strong> subgroups (Schiemann, 1989). Morerecently, Tsubokura et al. (1993) exp<strong>and</strong>ed the serogroups <strong>to</strong> 11 <strong>and</strong> also added asubgroup <strong>to</strong> O:1 (O:1C).Virulent strains of Y. pseudotuberculosis have a plasmid that determines the virulencefac<strong>to</strong>rs, including a kinase that determines the pathogenicity of the strains(Galyov et al., 1993).Geographic Distribution: The distribution of the etiologic agent is probablyworldwide. The greatest concentration of animal <strong>and</strong> human cases is found inEurope, the Russian Far East, <strong>and</strong> Japan.Occurrence in Man: For many years, pseudotuberculous yersiniosis was considereda disease that almost exclusively affected animals. However, since the 1950s,cases of lymphadenitis were described in children who had been operated on forappendicitis. In slightly more than three years, 117 cases of the disease werereported in Germany, most of which were diagnosed serologically. Hundreds ofcases were diagnosed in Europe in later years (Schiemann, 1989).Outbreaks occur as well as sporadic cases, which are possibly more numerous. Anepidemic outbreak with 19 cases occurred in Finl<strong>and</strong> (Tertti et al., 1984). In the


PSEUDOTUBERCULOUS YERSINIOSIS 219Russian Far East, a scarlatiniform form of the disease has been described, with severalthous<strong>and</strong> cases (S<strong>to</strong>vell, 1980). Three outbreaks occurred in the period1982–1984 in Okayama Prefecture (Japan). In one outbreak, serogroup 5a was isolatedfrom 16 patients <strong>and</strong> the infection was tied <strong>to</strong> contaminated foods. The othertwo outbreaks occurred in remote mountainous regions <strong>and</strong> affected a large numberof preschool- <strong>and</strong> school-aged children, as well as adults. In these two outbreaks, a<strong>common</strong> source of infection could not be found, although it may have been well orstream water. Serotype 2c was detected in the feces of one patient <strong>and</strong> in well water.In another case, serotype 4b was detected in the feces of the patient <strong>and</strong> of a wildanimal (Inoue et al., 1988).Also in Japan, outbreaks occurred in 1991 in Aomori Prefecture in four primaryschools <strong>and</strong> one secondary school. A <strong>to</strong>tal of 732 people became ill, including students,teachers, <strong>and</strong> administrative personnel; 134 were hospitalized. Y. pseudotuberculosisserotype 5a was isolated from 27 (81.8%) of the 33 samples examined.The strains isolated had the plasmid that determines various virulence fac<strong>to</strong>rs, suchas calcium dependence at 37°C <strong>and</strong> au<strong>to</strong>agglutination. The outbreak was attributed<strong>to</strong> food served in the schools, but no specific food could be pinpointed. The etiologicagent was also isolated from wastewater <strong>and</strong> the cooks’ feces (Toyokawa et al.,1993). Serotypes 1, 2, <strong>and</strong> 3 have been isolated in Asia, Europe, Canada, <strong>and</strong> the US;serotypes 4 <strong>and</strong> 5 have been isolated in Europe <strong>and</strong> Japan; <strong>and</strong> serotype 6 has beenisolated in a few cases in Japan (Quan et al., 1981).Occurrence in Animals: Numerous species of domestic <strong>and</strong> wild mammals,birds, <strong>and</strong> reptiles are naturally susceptible <strong>to</strong> the infection. The disease occurs sporadicallyin domestic animals. In Europe, devastating epizootics have been describedin hares. Epizootic outbreaks have occurred in guinea pigs, wild birds, turkeys,ducks, pigeons, <strong>and</strong> canaries. Serotype 1 predominates in animal disease.The Disease in Man: The disease mainly affects children, adolescents, <strong>and</strong> youngadults. In the past, the most recognized clinical form was mesenteric adenitis orpseudoappendicitis with acute abdominal pain in the right iliac fossa, fever, <strong>and</strong>vomiting. In the outbreaks in Okayama Prefecture, abdominal pains were accompaniedby diarrhea. In another large outbreak in Japan, 86.4% of 478 patients hadpyrexia, 73.8% had rashes, 66.7% had abdominal pain, <strong>and</strong> 63.4% experienced nausea<strong>and</strong> vomiting. Another frequent sign is strawberry <strong>to</strong>ngue <strong>and</strong> painful pharyngealredness. In the 19 patients studied in Finl<strong>and</strong> (Tertti et al., 1984), the diseaselasted from one week <strong>to</strong> six months. Twelve of the patients had complications: sixhad erythema nodosum, four had arthritis, one had iritis, <strong>and</strong> one had nephritis.The incubation period is still unclear, but is estimated <strong>to</strong> last from one <strong>to</strong>three weeks.Septicemia caused by Y. pseudotuberculosis is rare <strong>and</strong> usually appears in weakenedindividuals, particularly in the elderly or immunodeficient.In the Russian Far East, a scarlatiniform type of the disease has been described.This syndrome is characterized by fever, a scarlatiniform rash, <strong>and</strong> acute polyarthritis.The disease can be reproduced in volunteers using cultures of the agent isolatedfrom patients (S<strong>to</strong>vell, 1980).Y. pseudotuberculosis is sensitive <strong>to</strong> tetracycline. Ofloxacin proved very effectivein treatment tested on infected rats, but the beta-lactams were not effective(Lemaitre et al., 1991).


220 BACTERIOSESThe Disease in Animals: Outbreaks of yersiniosis in guinea pig colonies haveoccurred in several parts of the world with some frequency. The course of the diseasein these animals is usually subacute. The mesenteric lymph nodes becomeswollen <strong>and</strong> caseous, <strong>and</strong> sometimes there are nodular abscesses in the intestinalwall, spleen, liver, <strong>and</strong> other organs The animal rapidly loses weight <strong>and</strong> often hasdiarrhea. The disease lasts about a month. The septicemic form is rarer; the animaldies in a few days without showing significant symp<strong>to</strong>ms. Mortality varies from 5%<strong>to</strong> 75%. Apparently healthy animals infected with Y. pseudotuberculosis that remainin the colony can perpetuate the infection <strong>and</strong> cause new outbreaks. Serotype 1 wasisolated in an outbreak in a colony of guinea pigs in Argentina (Noseda et al., 1987).In cats, anorexia, gastroenteritis, jaundice, <strong>and</strong> often palpable mesenteric lymphnodes <strong>and</strong> hypertrophy of the spleen <strong>and</strong> liver are observed. Death can ensue two orthree weeks after the onset of the disease.Epizootics with abortions, suppurative epididymo-orchitis, <strong>and</strong> high mortalityhave been recorded in sheep in Australia <strong>and</strong> Europe. In Australian sheep, infectioncaused by serotype 3 of Y. pseudotuberculosis is <strong>common</strong> <strong>and</strong> occurs primarily inanimals 1 <strong>to</strong> 2 years of age. The infection lasts up <strong>to</strong> 14 weeks during winter <strong>and</strong>spring (Slee <strong>and</strong> Skilbeck, 1992). Affected animals usually experience diarrhea <strong>and</strong>weight loss. Symp<strong>to</strong>ms include characteristic microabscesses in the intestinalmucosa <strong>and</strong> increased thickness in the colonic <strong>and</strong> cecal mucosa (Slee <strong>and</strong> But<strong>to</strong>n,1990). Isolated cases with abortions <strong>and</strong> abscesses have been confirmed in sheep inseveral countries. Serotypes O:3 <strong>and</strong> O:1 have been isolated from goats in Australia.Diarrhea <strong>and</strong> loss of conditioning are the most notable symp<strong>to</strong>ms (Slee <strong>and</strong> But<strong>to</strong>n,1990). Abortions <strong>and</strong> neonatal death were described in a herd of goats (Witteet al., 1985).The infection <strong>and</strong> disease in cattle have been recognized in several countries. InAustralia, they are caused by serotype 3, which seems <strong>to</strong> prevail in the country’sruminants. In an episode of diarrhea in a dairy herd, 35 young animals died; in 20of 26 examined his<strong>to</strong>logically, the characteristic microabscesses were found in theintestinal mucosa. The disease occurred during the winter, spring, <strong>and</strong> early summer.In adult animals, there was a high rate of serologic reac<strong>to</strong>rs (Slee et al., 1988). Thedisease has also been described in Australia in adult cattle in flooded fields, withdiarrhea <strong>and</strong> death. Again, the serotype isolated was O:3 (Callinan et al., 1988).More recently, the disease was described in two herds in Argentina. In one herd,5.8% of the cattle became sick <strong>and</strong> 1.7% died. The symp<strong>to</strong>ms consisted of cachexia,diarrhea, <strong>and</strong> lack of mo<strong>to</strong>r coordination. In the second herd, 0.6% of 700 animalsdied <strong>and</strong> deaths occurred suddenly without prior symp<strong>to</strong>ms. The serotype responsiblewas also O:3 in Argentina (Noseda et al., 1990). In Canada, there have also beencases in cattle, with abortions <strong>and</strong> pneumonia.Cases of gastroenteritis have been observed in swine. Y. pseudotuberculosis hasbeen isolated from the feces <strong>and</strong> particularly from the <strong>to</strong>nsils of apparentlyhealthy animals.Outbreaks in turkeys have been described in the US (Oregon <strong>and</strong> California) <strong>and</strong>Engl<strong>and</strong>. An outbreak occurred on four farms in California (Wallner-Pendle<strong>to</strong>n <strong>and</strong>Cooper, 1983). The main symp<strong>to</strong>ms were anorexia; watery, yellowish-green diarrhea;depression; <strong>and</strong> acute locomo<strong>to</strong>r impairment. The disease affected males 9 <strong>to</strong>12 weeks old <strong>and</strong> had a morbidity rate of 2% <strong>to</strong> 15% <strong>and</strong> high mortality, principallydue <strong>to</strong> cannibalism. Administration of high doses of tetracyclines in food seemed <strong>to</strong>


PSEUDOTUBERCULOUS YERSINIOSIS 221arrest the disease, but the birds were condemned in the postmortem inspectionbecause of septicemic lesions. The principal lesions were necrotic foci in the liver<strong>and</strong> spleen, catarrhal enteritis, <strong>and</strong> osteomyelitis.The pseudotuberculosis agent is the most <strong>common</strong> cause of death in hares (Lepuseuropaeus) in France <strong>and</strong> Germany. Rabbits (Oryc<strong>to</strong>lagus cuniculus) <strong>and</strong> the ringdove(Columba palumbus) are also frequent victims of the disease. Epizootics havebeen described among rats (Rattus norvegicus) in Japan.In captive animals, disease caused by Y. pseudotuberculosis occurs with some frequency.Serotype O:1 was isolated in farm-bred nutrias (Myocas<strong>to</strong>r coypus); itaffected both young animals <strong>and</strong> adults with acute or chronic symp<strong>to</strong>ms. The principalsymp<strong>to</strong>ms were diarrhea, swelling of the lymph nodes, formation of nodulesin various organs, cachexia, <strong>and</strong> paralysis of the hindquarters (Cipolla et al., 1987;Monteavaro et al., 1990). In two London zoos, there were several deaths across abroad b<strong>and</strong> of mammalian <strong>and</strong> avian species. Disease <strong>and</strong> death occurred sporadically,particularly in winter. The most affected species was the Patagonian mara(Dolichotis patagonum). Death in captive animals due <strong>to</strong> Y. pseudotuberculosis represents0.66% <strong>to</strong> 0.79% of deaths each year. The serotypes isolated were 1a <strong>and</strong> 1b,which are the predominant types in many European countries. Some strains of 2awere also isolated (Parsons, 1991).The disease also occurs in captive monkeys. In one colony, one green monkey(Cercopithecus aethiops) <strong>and</strong> nine squirrel monkeys (Saimiri sciureus) became sick.The digestive system was most affected during the acute phase <strong>and</strong> the lymphatictissues, spleen, <strong>and</strong> liver suffered severe alteration in the chronic phase (Plesker <strong>and</strong>Claros, 1992). In another colony of New World monkeys, two different serotypeswere isolated (O:1 <strong>and</strong> O:2), depending on the group of origin (Brack <strong>and</strong>Gatesman, 1991; Brack <strong>and</strong> Hosefelder, 1992).Source of Infection <strong>and</strong> Mode of Transmission (Figure 16): Many facets of theepidemiology of pseudotuberculous yersiniosis still need <strong>to</strong> be clarified. The broadrange of animal <strong>and</strong> bird species that are naturally susceptible <strong>to</strong> the infection <strong>and</strong>are carriers of Y. pseudotuberculosis suggests that animals are the reservoir of theetiologic agent. In this enormous reservoir, researchers emphasize the role ofrodents <strong>and</strong> various bird species. In mountainous areas of Shimane Prefecture(Japan), a bacteriological study was conducted of 1,530 wild mice of the generaApodemus <strong>and</strong> Eothenomys, <strong>and</strong> moles (Urotrichus talpoides). Y. pseudotuberculosiswas isolated from the cecum of 72 animals <strong>and</strong> 10 of the strains had virulenceplasmids. The etiologic agent was detected only in the mice, more frequently duringthe mating season <strong>and</strong> in newborns (Fukushima et al., 1990). Another study in thesame prefecture cultured feces from 610 wild mammals <strong>and</strong> 259 wild birds. Thirtysevenstrains of Y. pseudotuberculosis were isolated from 34 mammals (5.6%) <strong>and</strong>from 2 anserine fowl (0.8%). The serotypes isolated were the same as those isolatedfrom humans in that region of Japan, thus the inference of an epidemiological connectionbetween the human infection <strong>and</strong> the infection in wild animals. The highestrate of infection (14%) was obtained in an omnivorous canine, the raccoon dog(Nyctereutes procyonoides), which is <strong>common</strong> in Japan, China, <strong>and</strong> Korea(Fukushima <strong>and</strong> Gomyoda, 1991).In studies conducted in Germany <strong>and</strong> Holl<strong>and</strong>, the agent was isolated from 5.8%<strong>and</strong> 4.3% of the <strong>to</strong>nsils of 480 <strong>and</strong> 163 clinically health swine, respectively, indi-


222 BACTERIOSESFigure 16. Pseudotuberculous yersiniosis (Yersinia pseudotuberculosis).Probable mode of transmission.Rodents,fowl, <strong>and</strong>lagomorphsElimination throughfeces <strong>and</strong> urineContaminated food<strong>and</strong> environmentIngestionRodents,fowl, <strong>and</strong>lagomorphsIngestion of contaminatedfood, contactMancating that this animal is a healthy carrier (Weber <strong>and</strong> Knapp, 1981a). In Japan, 2%of pig <strong>to</strong>ngues <strong>and</strong> 0.8% of chopped pork contained Y. pseudotuberculosis. Whensamples taken from retail pork were examined, two of the four strains isolated (corresponding<strong>to</strong> serotype 4b) had the same pathogenic properties as the human strainsobtained from patients (Shiozawa et al., 1988). One 4b strain was isolated previouslyfrom pork by Fukushima (1985). The agent was isolated in 0.58% of 1,206samples of swine feces examined over 14 months. These isolations, as well as thosefrom <strong>to</strong>nsils, were done in the cold months, corresponding <strong>to</strong> the season in whichhuman cases occur (Weber <strong>and</strong> Knapp, 1981a). In New Zeal<strong>and</strong>, the agent has frequentlybeen isolated from deer. A study conducted in cattle in the same country isolatedthe agent from 134 (26.3%) of 509 fecal samples from 84% of 50 herds.Serotype 3 was the most prevalent (93.2%), followed by 1 <strong>and</strong> 2. None of the herdshad prior his<strong>to</strong>ry of disease due <strong>to</strong> Y. pseudotuberculosis, <strong>and</strong> thus were healthy carriers.The study was conducted in young animals during the winter (Hodges <strong>and</strong>Carman, 1985). The authors note that diagnosis should not rely solely on examinationof feces.Several authors believe the soil is the reservoir of the agent, but isolations fromthe soil in Europe have primarily yielded serotype 2, which is rarely found in thehuman disease (Aldova et al., 1979). However, in the focus of scarlatiniformpseudotuberculosis in the Russian Far East, serotype 1 has been isolated from water<strong>and</strong> soil possibly contaminated by animal feces, which would explain the large numberof cases. In Khabarovsk Kray, in the Asiatic northeast of the Russian Federation,the disease changed seasons in the period 1983–1989, going from winter <strong>to</strong> the middleof summer. This change could be explained by the early provision of vegetablesin s<strong>to</strong>res, which could be contaminated by the feces of wild <strong>and</strong> synanthropic


PSEUDOTUBERCULOUS YERSINIOSIS 223Muridae (Dziubak et al., 1991). In any case, animals <strong>and</strong> wild fowl undoubtedlycontribute <strong>to</strong> environmental contamination. An epizootic or epornitic in one animalspecies often has repercussions in other species due <strong>to</strong> the excretion of the agent infeces <strong>and</strong> contamination of the environment.The mode of transmission is fecal-oral. The localization of the infection in themesenteric lymph nodes indicates that the digestive tract is the bacteria’s principalroute of entry.In repeated outbreaks of yersiniosis in guinea pig colonies in Great Britain, theinfection was transmitted by vegetables contaminated with feces of the ringdove(Columba palumbus). In the outbreak of pseudotuberculosis in turkeys in California(USA) (Wallner-Pendle<strong>to</strong>n <strong>and</strong> Cooper, 1983), two dead squirrels were found nearthe feeders. The etiologic agent was isolated from necrotic lesions in the liver <strong>and</strong>spleen of one of the squirrels. The immediate source of infection for man is oftendifficult <strong>to</strong> ascertain. A <strong>common</strong> source of infection was not found for the epidemicoutbreak in 19 patients in Finl<strong>and</strong> (Tertti et al., 1984).The vehicles of infection are pork <strong>and</strong> possibly meat from other species; waterfrom contaminated wells <strong>and</strong> streams; <strong>and</strong> vegetables contaminated by feces of wildanimals, rodents, <strong>and</strong> other mammals <strong>and</strong> birds.In both man <strong>and</strong> animals, the disease is prevalent in the cold months. Two reasonsare suggested for this phenomenon. The agent survives better at low temperatures<strong>and</strong> many animals are healthy carriers that become ill when stressed by cold, moisture,<strong>and</strong> poor nutrition, <strong>and</strong> eliminate the agent in their feces (Carniel <strong>and</strong> Mollaret,1990). Parturition is another stress fac<strong>to</strong>r. Young animals are more susceptible. Theinfection is transmitted from animal <strong>to</strong> animal in contaminated pastures.Role of Animals in the Epidemiology of the Disease: Wild mammals, rodents,<strong>and</strong> others, as well as domestic mammals (swine) <strong>and</strong> wild birds, constitute thereservoir. The most <strong>common</strong> route of transmission <strong>to</strong> man is perhaps indirectlythrough contamination of the environment <strong>and</strong> foods by feces. The agent can survivefor a relatively long time on vegetables <strong>and</strong> inanimate objects. A case of transmissionby dog bite is also known.Diagnosis: Definitive diagnosis can only be obtained through isolation <strong>and</strong> identificationof the causal agent. The most suitable material is the mesenteric lymphnodes. The agent can be isolated from contaminated samples in culture media usedfor enterobacteria. A selective agar called cefsulodin-irgasan-novobiocin (CIN) canbe used for epidemiological studies. Enrichment with diluted alkalis has been usedsuccessfully for isolations from meat samples (Fukushima, 1985). Serotyping of isolatedstrains is important from an epidemiological perspective. Serological tests<strong>common</strong>ly used <strong>to</strong> determine infection by Y. pseudotuberculosis are agglutination,hemagglutination, complement fixation, <strong>and</strong> more recently, enzyme-linkedimmunosorbent assay (ELISA) with the corresponding serotype, which is consideredmost sensitive <strong>and</strong> specific. Results should be carefully evaluated, since Y.pseudotuberculosis <strong>and</strong> Y. enterocolitica give cross-reactions <strong>and</strong> various serotypeshave antigens in <strong>common</strong> with other enterobacteria.Control: The principal preventive measure consists of protecting food <strong>and</strong> wateragainst fecal contamination by rodents <strong>and</strong> fowl. Controlling peridomestic rodentpopulations <strong>and</strong> limiting the number of birds in public places are also recommended.


224 BACTERIOSESMeats <strong>and</strong> other animal products should be well cooked. Only chlorinated watershould be consumed or, in its absence, water should be boiled for several minutes.Vegetables should be washed well with chlorinated water.BibliographyAldova, E., A. Brezinova, J. Sobotkova. A finding of Yersinia pseudotuberculosis in wellwater. Zbl Bakt Hyg [B] 169:265–270, 1979.Bercovier, H., H.H. Mollaret, J.M. Alonso, J. Brault, G.R. Fanning, A.G. Steigerwalt, et al.Intra <strong>and</strong> interspecies relatedness of Yersinia pestis by DNA hybridization <strong>and</strong> its relationship<strong>to</strong> Yersinia pseudotuberculosis. Curr Microbiol 4:225–229, 1980.Brack, M., T.J. Gatesman [Yersinia pseudotuberculosis in New World monkeys]. BerlMunch Tierarztl Wochenschr 104:4–7, 1991.Brack, M., F. Hosefelder. In vitro characteristics of Yersinia pseudotuberculosis of nonhumanprimate origin. Zentralbl Bakteriol 277:280–287, 1992.Callinan, R.B., R.W. Cook, J.G. Boul<strong>to</strong>n, et al. Enterocolitis in cattle associated withYersinia pseudotuberculosis infection. Aust Vet J 65:8–11, 1988.Carniel, E., H.H. Mollaret. Yersiniosis [review]. Comp Immunol Microbiol Infect Dis13:51–58, 1990.Cipolla, A.L., P.E. Martino, J.A. Villar, M. Catena. Rodenciosis en nutrias (Myocas<strong>to</strong>r coypus)de criadero: primeros hallazgos en Argentina. Rev Argent Prod Animal 7:481–486, 1987.Dziubak, V.F., A.S. Maramovich, I.I. Lysanov, R.N. Liberova. [The epidemiological patternsof pseudotuberculosis in Khabarovsk Kray]. Zh Mikrobiol Epidemiol Immunobiol.Oc<strong>to</strong>ber (10):25–28, 1991.Fukushima, H. Direct isolation of Yersinia enterocolitica <strong>and</strong> Yersinia pseudotuberculosisfrom meat. Appl Environ Microbiol 50:710–712, 1985.Fukushima, H., M. Gomyoda. Intestinal carriage of Yersinia pseudotuberculosis by wildbirds <strong>and</strong> mammals in Japan. Appl Environ Microbiol 57:1152–1155, 1991.Fukushima, H., M. Gomyoda, S. Kaneko. Mice <strong>and</strong> moles inhabiting mountainous areas ofShimane Peninsula as sources of infection with Yersinia pseudotuberculosis. J Clin Microbiol28:2448–2455, 1990.Galyov, E.E., S. Hakansson, A. Forsberg, H. Wolf-Watz. A secreted protein kinase ofYersinia pseudotuberculosis is an indispensable virulence determinant. Nature 361(6414):730–732, 1993.Hodges, R.T., M.G. Carman. Recovery of Yersinia pseudotuberculosis from faeces ofhealthy cattle. N Z Vet J 33:175–176, 1985.Inoue, M., H. Nakashima, T. Ishida, M. Tsubokura. Three outbreaks of Yersinia pseudotuberculosisinfection. Zbl Bakt Hyg [B] 186:504–511, 1988.Joubert, L. La pseudo-tuberculose, zoonose d’avenir. Rev Med Vet Lyon 119:311–322, 1968.Lemaitre, B.C., D.A. Mazigh, M.R. Scavizzi. Failure of beta-lactam antibiotics <strong>and</strong> markedefficacy of fluoroquinolones in treatment of murine Yersinia pseudotuberculosis infection.Antimicrob Agents Chemother 35:1785–1790, 1991.Mair, N.S. Yersiniosis in wildlife <strong>and</strong> its public health implications. J Wildl Dis 9:64–71, 1973.Mair, N.S. Yersiniosis (Infections due <strong>to</strong> Yersiniosis pseudotuberculosis <strong>and</strong> Yersiniosisenterocolitica). In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger, eds. DiseasesTransmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.Monteavaro, C., A. Schettino, P. So<strong>to</strong>, et al. Aislamien<strong>to</strong> de Yersinia pseudotuberculosis ennutrias de criadero. Rev Med Vet 71:220–224, 1990.


PSEUDOTUBERCULOUS YERSINIOSIS 225Noseda, R.P., J.C. Bardón, A.H. Martínez, J.M. Cordeviola. Yersinia pseudotuberculosis:epizootia en una colonia de Cavia porcellus. Vet Argent 4:134–136, 1987.Noseda, R.P., A.H. Martínez, J.C. Bardón, et al. Yersinia pseudotuberculosis en bovinos dela Provincia de Buenos Aires. Vet Argent 7:385–388, 1990.Parsons, R. Pseudotuberculosis at the zoological society of London (1981 <strong>to</strong> 1987). Vet Rec128:130–132, 1991.Plesker, R., M. Claros. A spontaneous Yersinia pseudotuberculosis infection in a monkeycolony. Zbl Vet Med [B] 39:201–208, 1992.Quan, T.J., A.M. Barnes, J.D. Pol<strong>and</strong>. Yersinioses. In: A. Balows, W.J. Hausler, Jr., eds.Diagnostic Procedures for Bacterial, Mycotic <strong>and</strong> Parasitic Infections. Washing<strong>to</strong>n, D.C.:American Public Health Association; 1981.Schiemann, D.A. Yersinia enterocolitica <strong>and</strong> Yersinia pseudotuberculosis. In: Doyle, M.P.Foodborne Bacterial Pathogens. New York: Marcel Dekker; 1989.Shiozawa, K., M. Hayashi, M. Akiyama, et al. Virulence of Yersinia pseudotuberculosisisolated from pork <strong>and</strong> from the throats of swine. Appl Environ Microbiol 54:818–821, 1988.Slee, K.J., P. Brightling, R.J. Seiler. Enteritis in cattle due <strong>to</strong> Yersinia pseudotuberculosisinfection. Aust Vet J 65:271–275, 1988.Slee, K.J., C. But<strong>to</strong>n. Enteritis in sheep, goats <strong>and</strong> pigs due <strong>to</strong> Yersinia pseudotuberculosisinfection. Aust Vet J 67:320–322, 1990.Slee, K.J., N.W. Skilbeck. Epidemiology of Yersinia pseudotuberculosis <strong>and</strong> Y. enterocoliticainfection in sheep in Australia. J Clin Microbiol 30:712–715, 1992.S<strong>to</strong>vell, P.L. Pseudotubercular yersiniosis. In: S<strong>to</strong>enner, H., W. Kaplan, M. Torten, eds. Vol2, Section A: CRC H<strong>and</strong>book Series in Zoonoses. Boca Ra<strong>to</strong>n: CRC Press; 1980.Tertti, R., K. Granfors, O.P. Leh<strong>to</strong>nen, J. Mertsola, A.L. Makela, I. Valimaki, et al. An outbreakof Yersinia pseudotuberculosis infection. J Infect Dis 149:245–250, 1984.Toyokawa, Y., Y. Oh<strong>to</strong>mo, T. Akiyama, et al. [Large scale outbreak of Yersinia pseudotuberculosisserotype 5a infection at Noheji-machi in Aomori Prefecture]. KansenshogakuZasshi 67:36–44, 1993.Tsubokura, M., S. Aleksic, H. Fukushima, et al. Characterization of Yersinia pseudotuberculosisserogroups O9, O10 <strong>and</strong> O11; subdivision of O1 serogroup in<strong>to</strong> O1a, O1b, <strong>and</strong> O1csubgroups. Zentralbl Bakteriol 278:500–509, 1993.Wallner-Pendle<strong>to</strong>n, E., G. Cooper. Several outbreaks of Yersinia pseudotuberculosis inCalifornia turkey flocks. Avian Dis 27:524–526, 1983.Weber, A., W. Knapp. [Seasonal isolation of Yersinia enterocolitica <strong>and</strong> Yersinia pseudotuberculosisfrom <strong>to</strong>nsils of healthy slaughter pigs]. Zbl Bakt Hyg A 250:78–83, 1981a.Weber, A., W. Knapp. [Demonstration of Yersinia enterocolitica <strong>and</strong> Yersinia pseudotuberculosisin fecal samples of healthy slaughter swine depending on the season]. Zbl Vet Med B28:407–413, 1981b.Wetzler, T.F. Pseudotuberculosis. In: Davis, J.W., L.H. Karstad, D.O. Trainer, eds.Infectious Diseases of Wild Mammals. Ames: Iowa State University Press; 1970.Witte, S.T., D.P. Sponenberg, T.C. Collins. Abortion <strong>and</strong> early neonatal death of kidsattributed <strong>to</strong> intrauterine Yersinia pseudotuberculosis infection. J Am Vet Med Assoc 187:834, 1985.


226 BACTERIOSESRAT-BITE FEVERICD-10 A25.0 spirillosis; A25.1 strep<strong>to</strong>bacillosisEtiology: Strep<strong>to</strong>bacillus moniliformis <strong>and</strong> Spirillum minus (S. minor).Rat-bite fever is caused by two different bacteria: Strep<strong>to</strong>bacillus moniliformis<strong>and</strong> Spirillum minus. Their geographic distribution <strong>and</strong> clinical picture are different<strong>and</strong> thus they will be treated separately.1. Infection due <strong>to</strong> Strep<strong>to</strong>bacillus moniliformisSynonyms: Haverhill fever, epidemic arthritic erythema, strep<strong>to</strong>bacillary fever.Etiology: Strep<strong>to</strong>bacillus moniliformis is a gram-negative, pleomorphous, nonmotile,nonsporogenic, microaerophilic bacillus 1 <strong>to</strong> 5 microns long <strong>and</strong> 0.1 <strong>to</strong> 0.7in diameter. It occurs in isolated form or in chains 10 <strong>to</strong> 150 microns long, dependingon the culture medium. Isolation of S. moniliformis requires media with a 20%supplement of serum, blood, or ascitic fluid (Savage, 1984).Geographic Distribution: Worldwide.Occurrence in Man: Very rare. It generally occurs in sporadic cases. Almost halfof all cases are due <strong>to</strong> bites from labora<strong>to</strong>ry rats. There have also been outbreaks inthe US <strong>and</strong> Great Britain. The name Haverhill fever derives from an outbreak of“epidemic arthritic erythema” that occurred in 1926 in Haverhill, Massachusetts(USA). The largest outbreak <strong>to</strong> date occurred in Great Britain. It affected 304 peopleat a girls’ school in a rural area, representing 43% of all the students <strong>and</strong> personnelat the school (McEvoy et al., 1987).Occurrence in Animals: The agent is isolated from the nasopharynx of a highpercentage of healthy rats. Epizootics have been described in wild <strong>and</strong> labora<strong>to</strong>rymice. There have been some outbreaks in turkeys <strong>and</strong> isolated cases in otheranimals.The Disease in Man: The incubation period lasts from 2 <strong>to</strong> 14 days after the bitefrom a rat or other rodent. The disease begins with symp<strong>to</strong>ma<strong>to</strong>logy similar <strong>to</strong> tha<strong>to</strong>f influenza: fever, headache, chills, <strong>and</strong> myalgia. The bite wound heals spontaneouslywithout complications. A maculopapular rash on the extremities as well asmigra<strong>to</strong>ry arthralgia <strong>and</strong> myalgia are <strong>common</strong>. Polyarthritis is seen in the mostsevere cases. After a short time, body temperature returns <strong>to</strong> normal, but the fevermay recur. Endocarditis is a possible complication. Mortality reaches 10% inuntreated cases.Haverhill fever has been attributed <strong>to</strong> the ingestion of milk contaminated by ratfeces. Its characteristics were the severity of vomiting <strong>and</strong> the incidence of pharyngitis,as well as the usual symp<strong>to</strong>ms of rat-bite fever (Washburn, 1990).The outbreak that affected so many people at the school in Great Britain wasattributed <strong>to</strong> water contaminated by rats. Many girls were hospitalized for weeks,with severe arthralgia <strong>and</strong> frequent relapses. There were also complications, such asendocarditis, pneumonia, metastatic abscesses, <strong>and</strong> anemia (McEvoy et al., 1987).


RAT-BITE FEVER 227The recommended treatment is intramuscular administration of penicillin for twoweeks. McEvoy et al. (1987) recommend treatment with erythromycin <strong>to</strong> preventthe spontaneous development of L forms during the disease.The Disease in Animals: Labora<strong>to</strong>ry <strong>and</strong> wild rats are healthy carriers <strong>and</strong> harborthe etiologic agent in their nasopharynx. Purulent lesions have sometimes beenobserved in these animals. S. moniliformis is pathogenic for rats <strong>and</strong> has producedepizootics among rats in labora<strong>to</strong>ries <strong>and</strong> in their natural habitat. In one epizooticamong labora<strong>to</strong>ry rats, high morbidity <strong>and</strong> mortality rates were recorded, with suchsymp<strong>to</strong>ms as polyarthritis, gangrene, <strong>and</strong> spontaneous amputation of members. Inguinea pigs, the agent can produce cervical lymphadenitis with large abscesses inthe regional lymph nodes. Some outbreaks have been described among turkeys inwhich the most salient symp<strong>to</strong>m was arthritis.Source of Infection <strong>and</strong> Mode of Transmission: Rats are the reservoir of theinfection. They harbor the etiologic agent in the nasopharynx <strong>and</strong> transmit it <strong>to</strong>humans by biting. In the Haverhill epidemic, the source was milk. According <strong>to</strong> theepidemiological investigation conducted on the school in Great Britain, the sourceof infection was drinking water contaminated by rat feces.All outbreaks are due <strong>to</strong> a <strong>common</strong> source, whereas sporadic cases are due <strong>to</strong> abite from a rat or other rodent. It would seem that man is not very susceptible, sincethere are very few recorded cases. Personnel working with labora<strong>to</strong>ry rodents areexposed <strong>to</strong> infection. People who live in rat-infested houses can become infectedwithout contact with rodents (Benenson, 1990). Infection among turkeys has beenattributed <strong>to</strong> rat bites. It is suspected that infection in mice <strong>and</strong> other rodents in labora<strong>to</strong>riescan be caused by aerosols when these rodents are kept in the same environmentas rats.Role of Animals in the Epidemiology of the Disease: Rats are the reservoir ofthe infection <strong>and</strong> play an essential epidemiological role.Diagnosis: Diagnosis is accomplished by isolating S. moniliformis from the bloodstreamor articular lesions in blood- or serum-enriched media. Inoculation of guineapigs or rats from colonies that are demonstrably free of infection can also be used.A few labora<strong>to</strong>ries use serological tests, such as tube agglutination, complementfixation, or immunofluorescence (Wilkins et al., 1988).Control: The principal means of prevention is control of the rat population. Otherimportant measures are pasteurization of milk <strong>and</strong> protection of food <strong>and</strong> wateragainst rodents. Labora<strong>to</strong>ry rats, mice, <strong>and</strong> guinea pigs should be kept in separateenvironments <strong>and</strong> personnel charged with their care should be instructed in properh<strong>and</strong>ling techniques.2. Infection due <strong>to</strong> Spirillum minusSynonyms: Sodoku, spirillary fever.Etiology: The etiologic agent is Spirillum minus. These bacteria are not well characterized<strong>and</strong> there are no reference strains because it is difficult <strong>to</strong> culture the spirillum.The genus name is still uncertain <strong>and</strong> the species name minor is considered


228 BACTERIOSESincorrect. It is a spiral-shaped bacterium with two or three twists; it is motile, 3 <strong>to</strong> 5microns long, <strong>and</strong> about 0.2 microns in diameter (Krieg, 1984).Geographic Distribution: Worldwide, but more frequent in the Far East.Occurrence in Man: Occasional.Occurrence in Animals: The incidence of the infection in rats varies in differentparts of the world. It affects 25% of rats in some regions.The Disease in Man: It is similar <strong>to</strong> the disease caused by S. moniliformis. Themost notable differences are that arthritic symp<strong>to</strong>ms are rare <strong>and</strong> that four weeksafter the bite there is a characteristic eruption with reddish or purple plaques. Theincubation period is one <strong>to</strong> four weeks. Fever begins suddenly <strong>and</strong> lasts a few days,but it recurs several times over a period of one <strong>to</strong> three months. There is a generalizedexanthema<strong>to</strong>us eruption that may reappear with each attack of fever. Althoughthe bite wound heals during the incubation period, it exhibits an edema<strong>to</strong>us infiltration<strong>and</strong> often ulcerates. Similarly, the lymph nodes become hypertrophic.Mortality is approximately 10% in untreated patients.Treatment consists of intramuscular administration of procaine penicillin fortwo weeks.The Disease in Animals: The infection is not apparent in rats.Source of Infection <strong>and</strong> Mode of Transmission: The reservoir is rats <strong>and</strong> otherrodents; their saliva is the source of infection for man. The infection is transmittedby bites.Role of Animals in the Epidemiology of the Disease: Rats play the principalrole. Human infections caused by bites from ferrets, dogs, cats, <strong>and</strong> other carnivoreshave also been described. It is presumed that these animals become contaminatedwhile catching rodents <strong>and</strong> thus act as mechanical transmitters.Diagnosis: Diagnosis is accomplished by dark-field microscopic examination ofinfiltrate from the wound, the lymph nodes, the erythema<strong>to</strong>us plaques, <strong>and</strong> from theblood. The most reliable diagnosis is obtained by intraperi<strong>to</strong>neal inoculation of micewith blood or infiltrate from the wound, followed by microscopic examination oftheir blood <strong>and</strong> peri<strong>to</strong>neal fluid some two weeks after inoculation. The bacteria donot grow in labora<strong>to</strong>ry culture media.Control: Control is based on reduction of the rat population <strong>and</strong> on constructionof rat-proof dwellings.BibliographyAnderson, L.C., S.L. Leary, P.J. Manning. Rat-bite fever in animal research labora<strong>to</strong>ry personnel.Lab Anim Sci 33:292–294, 1983.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bisseru, B. Diseases of Man Acquired from His Pets. London: Heinemann Medical; 1967.Boyer, C.I., D.W. Bruner, J.A. Brown. A Strep<strong>to</strong>bacillus, the cause of tendo-sheat infectionin turkeys. Avian Dis 2:418–427, 1958.


RHODOCOCCOSIS 229Krieg, N.R. Aerobic/microaerophilic, motil, helical/vibroid gram-negative bacteria. In:Krieg, N.R., J.G. Holt, eds. Vol. 1: Bergey’s Manual of Systematic Bacteriology. Baltimore:Williams & Wilkins; 1984.McEvoy, M.B., N.D. Noah, R. Pilsworth. Outbreak of fever caused by Strep<strong>to</strong>bacillusmoniliformis. Lancet 2:1361–1363, 1987.Ruys, A.C. Rat bite fevers. In: Van der Hoeden, J., ed. Zoonoses. Amsterdam:Elsevier; 1964.Savage, N. Genus Strep<strong>to</strong>bacillus. In: Krieg, N.R., J.G. Holt, eds. Vol. 1: Bergey’s Manualof Systematic Bacteriology. Baltimore: Williams & Wilkins; 1984.Washburn, R.G. Strep<strong>to</strong>bacillus moniliformis (Rat-bite fever). In: M<strong>and</strong>ell, G.L., R.G.Douglas, Jr., J.E. Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. NewYork: Churchill Livings<strong>to</strong>ne, Inc.; 1990.Wilkins, E.G.L., J.G.B. Millar, P.M. Cockroft, O.A. Okubadejo. Rat-bite fever in a gerbilbreeder. J Infect 16:177–180, 1988.Yamamo<strong>to</strong>, R., G.T. Clark. Strep<strong>to</strong>bacillus moniliformis infection in turkeys. Vet Rec79:95–100, 1966.RHODOCOCCOSISICD-10 J15.8 other bacterial pneumoniaEtiology: Rhodococcus (Corynebacterium) equi belongs <strong>to</strong> the orderActinomycetales; it has a coccoid or bacillary shape, is gram-positive, aerobic, nonmotile,encapsulated, <strong>and</strong> nonsporogenic. Its normal habitat is the soil; it is a saprophyticbacteria that requires few nutrients <strong>and</strong> multiplies abundantly in fecal matterfrom herbivores.Most strains of R. equi belong <strong>to</strong> 4 serogroups, which in turn contain 14 serotypes.Approximately 60% of the strains in North America belong <strong>to</strong> capsular serotype 1,<strong>and</strong> 26% belong <strong>to</strong> capsular serotype 2. In Japan, capsular serotype 3 predominatesin cultures isolated from foals (Timoney et al., 1988).R. equi is an opportunistic pathogen <strong>and</strong> is harbored in the macrophages in theanimal organism, causing a granuloma<strong>to</strong>us inflammation (Prescott, 1991). A 15- <strong>to</strong>17-kilodal<strong>to</strong>n antigen has been identified that is probably associated with the virulenceof R. equi (Takai et al., 1991a) <strong>and</strong> could be used as a marker for it.Geographic Distribution: Worldwide. Since 1923, when the first case ofrhodococcosis was described in foals in Switzerl<strong>and</strong>, the disease has been reportedon all continents. R. equi is frequently <strong>and</strong> abundantly isolated from soil where therehave been sick horses, but also from areas where there was no rhodococcosis, <strong>and</strong>even from soil where there have been no horses or other domestic animals recently(Bar<strong>to</strong>n <strong>and</strong> Hughes, 1980).Occurrence in Man: Very rare. From the first human case described in 1977 up<strong>to</strong> 1983, the literature records no more than 13 human cases (Van Etta et al., 1983).


230 BACTERIOSESCases are more frequent due <strong>to</strong> the AIDS epidemic <strong>and</strong> at least 20 more cases werereported from 1983 <strong>to</strong> 1990 (Prescott, 1991). In many parts of the world, particularlyin developing countries, physicians <strong>and</strong> hospital microbiologists know littleabout this disease. Consequently, underreporting is possible.Occurrence in Animals: Infection due <strong>to</strong> R. equi is recognized worldwide as animportant cause of bronchopneumonia, ulcerative enteritis, <strong>and</strong> lymphadenitis infoals, <strong>and</strong> less frequently in other animal species (Bar<strong>to</strong>n <strong>and</strong> Hughes, 1980).The Disease in Man: As in other animals, in man the lungs are the organ mos<strong>to</strong>ften affected. The disease appears with a fever lasting several days <strong>to</strong> severalweeks, discomfort, dyspnea, unproductive cough, <strong>and</strong>, frequently, chest pain.Initially, x-rays show infiltration with nodular lesions, particularly in the superiorlobes of the lungs. If the patient is not treated, the granuloma<strong>to</strong>us lesion can developin<strong>to</strong> suppuration <strong>and</strong> cavitation. Extrapulmonary cases, such as osteomyelitis, hemorrhagicdiarrhea <strong>and</strong> cachexia, pleurisy, abscesses, <strong>and</strong> lymphadenitis occur rarely(Prescott, 1991).The infection <strong>and</strong> the disease appear in immunocompromised patients. R. equi isan intracellular parasite of the macrophages, which explains the pyogranuloma<strong>to</strong>usnature of the disease <strong>and</strong> the predisposition of patients with cell-mediated immunesystem defects. Currently, AIDS patients represent 88% of cases. The remainingcases are patients undergoing immunosuppressive treatment due <strong>to</strong> neoplasias or anorgan transplant. HIV-infected patients have a higher incidence of simultaneoussecondary infections <strong>and</strong> higher mortality (54.5% as compared <strong>to</strong> 20% for patientsnot infected with HIV).Given the intracellular nature of rhodococcosis, the efficacy of the antimicrobialagent depends on its ability <strong>to</strong> penetrate the phagocytes. R. equi is sensitive <strong>to</strong> erythromycin,vancomycin, amikacin, gentamicin, neomycin, <strong>and</strong> rifampicin. Surgicalresection of the lesion or lesions is an important part of treatment (Prescott, 1991;Harvey <strong>and</strong> Sunstrum, 1991). The survival rate was 75% when antibiotic treatmentwas combined with surgical resection of the infected tissue. The survival rate forthose who received only antibiotics was 61% (Harvey <strong>and</strong> Sunstrum, 1991).The Disease in Animals: Rhodococcosis is a disease that occurs primarily in foalsfrom 2 <strong>to</strong> 6 months of age, <strong>and</strong> particularly from 2 <strong>to</strong> 4 months of age. This susceptibilityof young foals could be because at that age the passive immunity conferredby the mother is in decline <strong>and</strong> the animal’s own immune system is still immature.Foals older than 6 months are resistant, unless they have a defect in cellular immunityor another concurrent disease with a debilitating effect (Yager, 1987).Equine rhodococcosis appears as a subacute or chronic suppurative bronchopneumonia.Formation of abscesses is extensive, accompanied by a suppurativelymphadenitis. The lesions progress slowly. The degeneration of the macrophagescoincides with the lysis of pulmonary parenchyma. Formation of abscesses continueswith expansion of the purulent center. The infection is spread through the lymphaticsystem <strong>and</strong> affects the regional lymph nodes. Despite bacteremia no lesionsare found in the liver or spleen, which would indicate that fixed macrophages coulddestroy R. equi in the circula<strong>to</strong>ry system. It is estimated that approximately 50% offoals with bronchopneumonia develop concomitant ulcerative colitis <strong>and</strong> typhlitis. Asmall number of foals develop only intestinal lesions (Yager, 1987).


RHODOCOCCOSIS 231In infection caused by R. equi, we find both subclinical cases discovered uponau<strong>to</strong>psy <strong>and</strong> a disease with a fatal outcome in less than one week (26% of 89 foalswho died from rhodococcosis).The disease usually begins with a fever, rapid breathing, <strong>and</strong> cough <strong>and</strong> thenbecomes more intense. Mucopurulent nasal discharge <strong>and</strong> dyspnea are also <strong>common</strong>.Most cases occur in summer, when there are more foals at a susceptible age <strong>and</strong>the temperature favors growth of the bacteria.The recommended treatment is a combination of erythromycin <strong>and</strong> rifampicin,which have a synergistic effect, for 4 <strong>to</strong> 10 weeks. The two drugs are liposoluble <strong>and</strong>can penetrate the phagocytes. In the case of diarrhea, fluids <strong>and</strong> electrolytes shouldbe replaced.In swine, rhodococcosis appears as cervical <strong>and</strong> submaxillary lymphadenitis. R.equi has also been isolated from normal lymph nodes. Infection is rare in otherspecies. Some sporadic cases have been reported in cattle, goats, sheep, reptiles, <strong>and</strong>cats. In cattle, the few cases reported were pyometra, chronic pneumonia, <strong>and</strong> lymphadenitis(Bar<strong>to</strong>n <strong>and</strong> Hughes, 1980).Source of Infection <strong>and</strong> Mode of Transmission: R. equi is a saprophyte in soil.Its concentration depends on the presence of horses <strong>and</strong> ambient temperature. Fecesof herbivores greatly favor their growth. It is believed that one of the feces’ components,acetic acid, is the principal fac<strong>to</strong>r in the agent’s multiplication (Fraser et al.,1991). The prevalence of virulent R. equi (isolated from the soil <strong>and</strong> the feces offoals) on a horse-breeding farm where rhodococcosis is endemic is much higherthan on a farm that has no his<strong>to</strong>ry of the disease. Foals bred on an endemic farm areconstantly exposed <strong>to</strong> virulent strains of R. equi (Takai et al., 1991b). In NewZeal<strong>and</strong>, samples of fecal matter from different animals <strong>and</strong> from the environmenthave been examined. The most frequent isolates came from the feces of foals (82%),mares (76%), deer (89%), sheep (97%), goats (83%), pigeons (64%), <strong>and</strong> soil samples(94%) (Carman <strong>and</strong> Hodges, 1987). However, R. equi could only be isolatedfrom 2 of 521 human fecal samples (Mutimer et al., 1979).The route of infection for man is through inhalation. The gastroenteritis caused by R.equi that a few patients suffer from may be caused by swallowing sputum. A possibleanimal source of infection was assumed in 12 of 32 human patients (Prescott, 1991).The airborne route is also preponderant in foals that inhale dust from the soil. Incontrast, in swine the route of infection is probably oral, as indicated by their lesions(cervical <strong>and</strong> submaxillary lymphangitis). R. equi colonizes in the intestine of foalsin the first 2 months of life. The formation of antibodies <strong>and</strong> the increased rate offormation would indicate a subclinical infection, acquired orally (Takai et al., 1986;Hietala et al., 1985; Yager, 1987).Role of Animals in the Epidemiology of the Disease: Although many non–HIVinfectedpatients suffering from rhodococcosis acknowledged some exposure <strong>to</strong> animals,it is still unclear whether animals represent a source of infection for man.Herbivores contribute with their feces <strong>to</strong> the rapid multiplication of R. equi in thewarm months <strong>and</strong> sick foals seem <strong>to</strong> be responsible for spreading virulent strains.The reservoir of R. equi is the soil.Diagnosis: Positive diagnosis can be obtained by isolating the etiologic agent. R.equi grows in <strong>common</strong> labora<strong>to</strong>ry media. Sputum can be used for isolation, but it is


232 BACTERIOSESmuch more accurate <strong>to</strong> collect material from a bronchial sample obtained throughpercutaneous thoracic aspiration, or biopsy during a lobec<strong>to</strong>my. R. equi can sometimesbe found in cultures with a variety of other bacteria <strong>and</strong> may be inadvertentlydiscarded as “diphtheroid.” The etiologic agent could be isolated from the blood ofapproximately one-third of human patients with pneumonia (Prescott, 1991).Control: There are no practical measures for protecting humans or foals. It ismore reasonable <strong>to</strong> prevent <strong>diseases</strong> that predispose humans <strong>to</strong> infection by R. equi,particularly AIDS. Another measure could be <strong>to</strong> reduce the dose of immunosuppressivemedications whenever possible.There are no preventive vaccines for equine rhodococcosis. On horse-breedingfarms, the accumulation of feces <strong>and</strong> resulting multiplication of R. equi should notbe permitted. Dusty conditions should be avoided in <strong>and</strong> around stables. On endemicfarms, it is recommended that foals be examined frequently in the first months oflife <strong>and</strong> that sick foals be treated (Fraser et al., 1991).BibliographyBar<strong>to</strong>n, M.D., K.L. Hughes. Corynebacterium equi:A review. Vet Bull 50:65–80, 1980.Carman, M.G., R.T. Hodges. Distribution of Rhodococcus equi in animals, birds <strong>and</strong> fromthe environment. N Z Vet J 35:114–115, 1987.Fraser, C.M., J.A. Bergeron, A. Mays, S.E. Aiello, eds. The Merck Veterinary Manual. 7thed. Rahway: Merck; 1991.Harvey, R.L., J.C. Sunstrum. Rhodococcus equi infection in patients with <strong>and</strong> withouthuman immunodeficiency virus infection. Rev Infect Dis 13:139–145, 1991.Hietala, S.K., A.A. Ardans, A. Sansome. Detection of Corynebacterium equi-specific antibodyin horses by enzyme-linked immunosorbent assay. Am J Vet Res 46:13–15, 1985.Mutimer, M.D., J.B. Woolcock, B.R. Sturgess. Corynebacterium equi in human faeces.Med J Aust 2:422, 1979. Cited in: Prescott, J.F. Rhodococcus equi: An animal <strong>and</strong> humanpathogen. Clin Microbiol Rev 4:20–34, 1991.Prescott, J.F. Rhodococcus equi: An animal <strong>and</strong> human pathogen. Clin Microbiol Rev4:20–34, 1991.Takai, S., K. Koike, S. Ohbushi, et al. Identification of 15- <strong>to</strong> 17-kilodal<strong>to</strong>n antigens associatedwith virulent Rhodococcus equi. J Clin Microbiol 29:439–443, 1991a.Takai, S., S. Ohbushi, K. Koike, et al. Prevalence of virulent Rhodococcus equi in isolatesfrom soil <strong>and</strong> feces of horses from horse-breeding farms with <strong>and</strong> without endemic infections.J Clin Microbiol 29:2887–2889, 1991b.Takai, S., H. Ohkura, Y. Watanabe, S. Tsubaki. Quantitative aspects of fecal Rhodococcus(Corynebacterium) equi in foals. J Clin Microbiol 23:794–796, 1986.Timoney, J.F., J.H. Gillespie, F.W. Scott, J.E. Barlough. Hagan <strong>and</strong> Bruner’s Microbiology<strong>and</strong> Infectious Diseases of Domestic Animals. 8th ed. Ithaca: Coms<strong>to</strong>ck; 1988.Van Etta, L.L., G.A. Filice, R.M. Ferguson, D.N. Gerding. Corynebacterium equi:A reviewof 12 cases of human infection. Rev Infect Dis 5:1012–1018, 1983.Yager, J.A. The pathogenesis of Rhodococcus equi pneumonia in foals. Vet Microbiol14:225–232, 1987.


SALMONELLOSIS 233SALMONELLOSISICD-10 A02.0 salmonella enteritis; A02.1 salmonella septicaemia;A02.8 other specified salmonella infectionsSynonyms: Nontyphoid salmonellosis.Etiology: The genus Salmonella belongs <strong>to</strong> the family Enterobacteriaceae. It ismade up of gram-negative, motile (with a few exceptions), facultatively anaerobicbacteria. Salmonellae grow between 8°C <strong>and</strong> 45°C <strong>and</strong> at a pH of 4 <strong>to</strong> 8. They donot survive at temperatures higher than 70°C. Pasteurization at 71.1°C for 15 secondsis sufficient <strong>to</strong> destroy salmonellae in milk.These bacteria can resist dehydration for a very long time, both in feces <strong>and</strong> infoods for human <strong>and</strong> animal consumption. In addition, they can survive for severalmonths in brine with 20% salinity, particularly in products with a high protein or fatcontent, such as salted sausages; they also resist smoking. It has been indicated thatthey can survive for a long time in soil <strong>and</strong> water (<strong>WHO</strong> Expert Committee onSalmonellosis Control, 1988).A study conducted in Great Britain showed that S. typhimurium can survive 4 <strong>to</strong>14 months in the environment of facilities with infected calves, an important epidemiologicalfac<strong>to</strong>r (McLaren <strong>and</strong> Wray, 1991). It can survive in ripening cheddarcheese for 10 months at 7°C (el-Gazzar <strong>and</strong> Marth, 1992).Le Minor <strong>and</strong> Popoff (1987) used DNA:DNA hybridization <strong>to</strong> show that they aregenetically a single species. Various classification schemes have been proposed,leading <strong>to</strong> controversy <strong>and</strong> confusion. At present, the trend is <strong>to</strong> return <strong>to</strong> the schemeconceived by Kauffmann-White due <strong>to</strong> its simplicity <strong>and</strong> because it is clearer <strong>and</strong>more useful from a clinical <strong>and</strong> epidemiological st<strong>and</strong>point. The nomenclaturescheme of Edwards <strong>and</strong> Ewing that was frequently used, particularly in theAmericas, is being ab<strong>and</strong>oned (Farmer et al., 1984). As a result, the serotype termis used directly as a species. Thus, S. enterica serotype Typhimurium according <strong>to</strong>one scheme or Salmonella subspecies I serotype typhimurium according <strong>to</strong> anotherscheme would currently be S. typhimurium.The Kauffmann-White scheme divides salmonellae in<strong>to</strong> serotypes. O somatic, Hflagellar, <strong>and</strong> Vi capsular antigens are distinguished primarily on the basis of theirantigenic structure. Currently, there are close <strong>to</strong> 2,200 serotypes.Some serotypes have several different phenotypes, <strong>and</strong> their identification can beimportant in epidemiologic investigation. For example, biochemical tests were able<strong>to</strong> differentiate three biotypes of S. typhimurium, each of which was associated witha geographic <strong>and</strong> ecological region. S. gallinarum <strong>and</strong> S. pullorum are two nonmotilesalmonellae adapted <strong>to</strong> birds. Some authors consider them a single species orserotype because they are antigenically identical. However, each of these serotypescauses a different disease (fowl typhoid <strong>and</strong> pullorum disease). They can be distinguishedbecause, unlike S. gallinarum, S. pullorum does not use dulci<strong>to</strong>l or d-tartrate(D’Aoust, 1989).Phage typing is also useful for some serotypes. The Scottish SalmonellaReference Labora<strong>to</strong>ry studied 2,010 cultures of S. typhimurium <strong>and</strong> differentiated137 different groups of phage types/biotypes. Four major epidemic clones were recognizedthat accounted for 52% of the cultures, with a predominance of bovine <strong>and</strong>


234 BACTERIOSEShuman strains. Epidemiological investigation shows that most salmonellosis outbreakscaused by S. typhimurium were caused by a lysotype/biotype that remainedstable throughout the course of the epidemic (Barker et al., 1980). Plasmid profiles<strong>and</strong> patterns of antibiotic resistance are also useful as epidemic markers.Except for serotypes S. typhi <strong>and</strong> S. paratyphi A, <strong>and</strong> S. paratyphi C, which arestrictly human <strong>and</strong> whose only reservoir is man, all serotypes can be consideredzoonotic or potentially zoonotic.Salmonellae have several virulence fac<strong>to</strong>rs that contribute <strong>to</strong> causing diarrhea,bacteremia, <strong>and</strong> septicemia. These fac<strong>to</strong>rs include the lipopolysaccharide of theouter wall, pili, flagella, cy<strong>to</strong><strong>to</strong>xin, <strong>and</strong> entero<strong>to</strong>xin (Murray, 1986).Geographic Distribution: Worldwide. S. enteritidis is the most prevalentspecies, followed by S. typhimurium. Changes in the relative frequency of serotypescan be observed over short periods of time, sometimes within one or two years.Only a limited number of serotypes is isolated from man or animals in a singleregion or country. The predominance of one or another can vary over time. Someserotypes, such as S. enteritidis <strong>and</strong> S. typhimurium, are found worldwide; in contrast,S. weltevreden seems <strong>to</strong> be confined <strong>to</strong> Asia.Occurrence in Man: It is very <strong>common</strong>. Salmonellosis occurs both in sporadiccases <strong>and</strong> outbreaks affecting a family or several hundreds or thous<strong>and</strong>s of people ina population. The true incidence is difficult <strong>to</strong> evaluate, since many countries do nothave an epidemiological surveillance system in place, <strong>and</strong> even where a system doesexist, mild <strong>and</strong> sporadic cases are not usually reported. In countries with a reportingsystem, the number of outbreaks has increased considerably in recent years; thisincrease is in part real <strong>and</strong> in part due <strong>to</strong> better reporting.In 1980, Salmonella was isolated from slightly more than 30,000 people in the US(CDC, 1982). In 1986, 42,028 cases were isolated (Hargrett-Bean et al., 1988). Inthe US <strong>and</strong> many other countries, the prevalent serotype was S. typhimurium. From1976 <strong>to</strong> 1993, the rate of isolation of S. enteritidis increased (21% of all isolates) <strong>and</strong>over<strong>to</strong>ok S. typhimurium as the most <strong>common</strong> serotype. During the period1985–1991, 375 outbreaks caused by S. enteritidis were reported, with 12,784 cases,1,508 hospitalized cases, <strong>and</strong> 49 deaths. Most of the cases were sporadic or smallfamily outbreaks, <strong>and</strong> many of them were from the same phage type, indicating thepossibility of a single source of infection (CDC, 1992a).During a conference held in 1990 that was attended by 1,900 people from 30states in the US, at least 23% became ill with gastroenteritis caused by S. enteritidis.The source of infection was a dessert prepared with eggs that were possibly undercooked(CDC, 1990).In 1985, an outbreak occurred in Illinois (USA) that affected 20,000 people <strong>and</strong>was caused by pasteurized milk contaminated by S. typhimurium that was multiresistant<strong>to</strong> antibiotics (ampicillin <strong>and</strong> tetracycline).Table 3 shows information on some outbreaks in the period 1981–1985 in variouscountries (<strong>WHO</strong> Expert Committee on Salmonellosis Control, 1988).According <strong>to</strong> several authors’ estimates, the number of human cases occurringeach year in the US ranges from 740,000 <strong>to</strong> 5,300,000. In Canada, the data weresimilar (Bryan, 1981). Rates for reported cases are about 10 per 100,000 inhabitantsin Denmark, 44 per 100,000 in Finl<strong>and</strong>, <strong>and</strong> 43 per 100,000 in Sweden, one-third <strong>to</strong>two-thirds of which were probably contracted by international travelers (Silliker,


SALMONELLOSIS 235Table 3. Outbreaks of foodborne salmonellosis in selected countries, 1981–1985.Approx. numberYear Country Food Serotype of cases1981 Engl<strong>and</strong> Chicken montevideo 5001981 The Netherl<strong>and</strong>s Cold buffet indiana 7001981 Scotl<strong>and</strong> Raw milk typhimurium 6541982 Engl<strong>and</strong> Chocolate napoli 2451982 Norway Black pepper oranienburg 1261984 Engl<strong>and</strong> Cooked meat virchow 2741984 Engl<strong>and</strong> Cold roast beef typhimurium 4501984 Canada Cheddar cheese typhimurium 2,0001984 Worldwide a Meat gelatin enteritidis 7661985 Engl<strong>and</strong> Cooked meats infantis 1501985 Engl<strong>and</strong> Powdered milk ealing 60for infants1985 United States Pasteurized milk typhimurium 20,000of AmericaaMeals prepared in London for airlines.Source: World Health Organization, 1988.1982). In the former West Germany, 33,215 cases were reported in 1978, 40,717 in1979, <strong>and</strong> 48,607 in 1980 (Poehn, 1982). In Australia, from 1980 <strong>to</strong> 1983, annualincidence was 32.2 per 100,000 inhabitants <strong>and</strong> in 1985 it was 27.0 per 100,000(D’Aoust, 1989).Seven of the 23 outbreaks of gastroenteritis that occurred on transatlantic flights<strong>to</strong> the US between 1947 <strong>and</strong> 1984 were due <strong>to</strong> salmonellosis (Tauxe et al., 1987).It is difficult <strong>to</strong> evaluate the situation of this disease in developing countriesbecause of the lack of epidemiological surveillance data, but epidemic outbreaks areknown <strong>to</strong> occur. In 1977, an extensive outbreak <strong>to</strong>ok place in Trujillo (Peru) amonguniversity students who lunched in the university dining hall. Of 640 students whoate regularly in the hall, 598 (93%) became ill <strong>and</strong> 545 were hospitalized, resultingin temporary overcrowding of community medical services. Serotype S. thompsonwas isolated from the patients’ s<strong>to</strong>ols, <strong>and</strong> epidemiologic evidence pointed <strong>to</strong> eggsused in the food as the source of the infection (Gunn <strong>and</strong> Bullón, 1979). In theperiod 1969–1974, 3,429 cases of acute diarrhea in children in Buenos Aires(Argentina) <strong>and</strong> its environs were studied. Isolations of 932 strains of Salmonellawere obtained from 3,429 s<strong>to</strong>ol samples. Between 1969 <strong>and</strong> 1972, isolation of S.typhimurium predominated, revealing the existence of an epidemic. The clinical picturewas serious <strong>and</strong> the mortality rate reached 14% of the 246 children studied.After 1972, isolations of serotype S. oranienburg increased, <strong>and</strong> those of S.typhimurium decreased. Seventy-three percent of the children acquired the infectionat home; 27% first showed symp<strong>to</strong>ms in the hospital after being admitted for causesother than gastrointestinal disorders (Binsztein et al., 1982). Starting in 1986, therewas a notable increase in S. enteritidis in Europe, the US, South America, <strong>and</strong> someAfrican countries. From 1986 <strong>to</strong> 1988, 39 outbreaks occurred in Argentina, affectingmore than 2,500 people, with a serious clinical picture (Eiguer et al., 1990;Rodrigue et al., 1990).


236 BACTERIOSESOccurrence in Animals: It is very <strong>common</strong>. The rate of infection in domesticanimals has been estimated at from 1% <strong>to</strong> 3%. In 1980, 16,274 strains of 183serotypes of Salmonella were isolated in the former West Germany from animals,foods of animal origin, water, <strong>and</strong> other sources (Pietzsch, 1982). In 1985,Salmonella was isolated from 1.25% of 222,160 samples of meat obtained duringveterinary inspection in slaughterhouses. In other examinations of animals <strong>and</strong> animalorgans, Salmonella was isolated from 4.81% of 81,851 examined. Positive cultureswere obtained from 4.59% of 141,827 bovine fecal samples. In the US, 2,515cultures of nonhuman origin were obtained in 1980 (CDC, 1982). Several surveyshave found the incidence of avian salmonellosis <strong>to</strong> be lower than 1% in Sweden,approximately 5% in Denmark, <strong>and</strong> approximately 7% in Finl<strong>and</strong>. Its incidence inother countries is higher. In Great Britain, there were 3,626 isolations in 1980 <strong>and</strong>2,992 in 1981. Epidemiologic surveillance of animals, including birds, is of theutmost importance, since the source of the large majority of nontyphoid salmonellosiscases is food of animal origin. There are no data from developing countries inthis regard.Some reports on pets indicate that salmonellosis occurs frequently. In the formerWest Germany between 1967 <strong>and</strong> 1983, different researchers isolated salmonellaefrom 8.4% <strong>to</strong> 12.8% of the dogs examined <strong>and</strong> from 9.8% <strong>to</strong> 11.2% of 908 cats. Inthe US, 0.04% of 124,774 dogs examined during the same time period gave positivecultures, as did 0.1% of 29,613 cats. In Iran, 7.7% of 672 dogs <strong>and</strong> 13.6% of 301cats were positive (D’Aoust, 1989).Infection caused by Salmonella is also spread among wild mammals, birds,amphibians, reptiles, <strong>and</strong> invertebrates.The Disease in Man: With the exception of S. typhi <strong>and</strong> the paratyphoidserotypes (particularly A <strong>and</strong> C), which are species-specific for man, all other infectionscaused by Salmonella may be considered <strong>zoonoses</strong>. Salmonellosis is perhapsthe most widespread zoonosis in the world.Salmonellae of animal origin cause an intestinal infection in man characterized bya 6- <strong>to</strong> 72-hour incubation period after ingestion of the implicated food, <strong>and</strong> suddenonset of fever, myalgias, cephalalgia, <strong>and</strong> malaise. The main symp<strong>to</strong>ms consist ofabdominal pain, nausea, vomiting, <strong>and</strong> diarrhea. Salmonellosis normally has abenign course <strong>and</strong> clinical recovery ensues in two <strong>to</strong> four days. The convalescentcarrier may shed salmonellae for several weeks <strong>and</strong>, more rarely, for a few months.Conversely, the carrier state is persistent in infections due <strong>to</strong> S. typhi or paratyphoidsalmonellae. Although salmonellosis may occur in persons of all ages, incidence ismuch higher among children <strong>and</strong> the elderly. Dehydration can be serious.Extraintestinal infections caused by zoonotic salmonellae are relatively infrequent.Of the 6,564 strains of Salmonella spp. isolated from 1969 <strong>to</strong> 1983 in a hospitalin Liverpool (Engl<strong>and</strong>), 3% were extraintestinal infections. Of the 194 extraintestinalcultures, 34% were from blood, 32% from urine, 23% from pus <strong>and</strong>inflamed tissues, 5% from bones, 5% from cerebrospinal fluid, <strong>and</strong> 3% from sputum(Wilkins <strong>and</strong> Roberts, 1988).Serotypes adapted <strong>to</strong> a particular animal species are usually less pathogenic forman (pullorum, gallinarum, abortus equi, abortus ovis). An exception is S. choleraesuis,which produces a serious disease with a septicemic syndrome,splenomegaly, <strong>and</strong> high fever a few days <strong>to</strong> a few weeks after the onset of gastroen-


SALMONELLOSIS 237teritis. Bacteremia is present in more than 50% of patients with S. choleraesuisinfections <strong>and</strong> the fatality rate may reach 20%. Serotypes sendai <strong>and</strong> dublin can alsocause septicemia (“enteric fever”) <strong>and</strong> often metastatic abscesses.Zoonotic salmonellae usually heal without complications <strong>and</strong> the only treatmentrecommended is rehydration <strong>and</strong> electrolyte replacement. A small proportion ofpatients, particularly those weakened by other <strong>diseases</strong> (AIDS, neoplasias, diabetes,etc.), can suffer from bacteremia. There may also be different localizations, such asthe lungs, pleura, joints, <strong>and</strong>, more rarely, the endocardium. Children under the ageof 5 <strong>and</strong> the elderly are more susceptible <strong>to</strong> complications. Children younger than 2months, the elderly, <strong>and</strong> patients with concurrent <strong>diseases</strong> should be given antibiotics(ampicillin, amoxycillin, cotrimoxazole, <strong>and</strong> chloramphenicol). Antibioticsshould also be given <strong>to</strong> patients with a prolonged fever with extraintestinal complications(Benenson, 1990).A high proportion of Salmonella strains with multiple antibiotic resistance hasbeen seen in many countries. The main cause of this in industrialized countries hasbeen the overuse of antibiotics in animal feed as a growth enhancer, as well as theindiscriminate prescription-drug treatment of people <strong>and</strong> animals. In Great Britain,the prophylactic use of antibiotics against bovine salmonellosis has resulted in theemergence of multiresistant strains of S. typhimurium,which have caused epizooticswith high mortality. Outbreaks <strong>and</strong> epidemics of multiresistant strains of severalserotypes have occurred in nurseries <strong>and</strong> pediatric clinics, with complications ofsepticemia or meningitis <strong>and</strong> high mortality. An epidemic caused by multiresistantstrains of serotype wien originated in Algeria in 1969 <strong>and</strong> spread <strong>to</strong> severalEuropean <strong>and</strong> Asian countries; the source in the food chain was not discovered.Other epidemics spreading <strong>to</strong> several countries have been caused by S. typhimuriumphage type 208 (<strong>WHO</strong> Scientific Working Group, 1980) <strong>and</strong>, in more recent years,by S. enteritidis. In developing countries, the principal cause of the emergence ofmultiresistant Salmonella strains may be self-medication, made possible by the public’seasy access <strong>to</strong> antibiotics without a prescription.The Disease in Animals: Salmonellae have a wide variety of domestic <strong>and</strong> wildanimal hosts. The infection may or may not be clinically apparent. In the subclinicalform, the animal may have a latent infection <strong>and</strong> harbor the pathogen in itslymph nodes, or it may be a carrier <strong>and</strong> eliminate the agent in its fecal materialbriefly, intermittently, or persistently. In domestic animals, there are several wellknownclinical entities due <strong>to</strong> species-adapted serotypes, such as S. pullorum or S.abortus equi. Other clinically apparent or inapparent infections are caused byserotypes with multiple hosts.CATTLE: The principal causes of clinical salmonellosis in cattle are serotypedublin <strong>and</strong> S. typhimurium. Other serotypes can sometimes be isolated fromsick animals.Salmonellosis in adult cattle occurs sporadically, but in calves it usually acquiresepizootic proportions. The disease generally occurs when stress fac<strong>to</strong>rs are involved.Serotype dublin, adapted <strong>to</strong> cattle, has a focal geographic distribution. In theAmericas, outbreaks have been confirmed in the western United States, Venezuela,Brazil, <strong>and</strong> Argentina. It also occurs in Europe <strong>and</strong> South Africa.In adult cattle, the disease begins with high fever <strong>and</strong> the appearance of bloodclots in the feces, followed by profuse diarrhea, <strong>and</strong> then a drop in body tempera-


238 BACTERIOSESture <strong>to</strong> normal. Signs of abdominal pain are very pronounced. Abortion is <strong>common</strong>.The disease may be fatal within a few days or the animal may recover, in which caseit often becomes a carrier <strong>and</strong> new cases appear. Calves are more susceptible thanadults, <strong>and</strong> in them the infection gives rise <strong>to</strong> true epidemic outbreaks, often withhigh mortality. Septicemia <strong>and</strong> death are frequent in newborns. The carrier state isless frequent among young animals <strong>and</strong> occurs primarily in adult cattle. The infectionis almost always spread by the feces of a cow that is shedding the agent, but itmay also originate from milk.SWINE: Swine are host <strong>to</strong> numerous Salmonella serotypes <strong>and</strong> are the principalreservoir of S. choleraesuis. Serotypes that attack swine include S. enteritidis, S.typhimurium, <strong>and</strong> S. dublin. These serotypes are generally isolated from the intestine<strong>and</strong> from the mesenteric lymph nodes. S. choleraesuis is very invasive <strong>and</strong>causes septicemia; it may be isolated from the blood or from any organ. Swine areparticularly susceptible <strong>and</strong> experience epidemic outbreaks between 2 <strong>and</strong> 4months of age, but the infection also appears in mature animals, almost always asisolated cases.Swine paratyphoid (S. choleraesuis) or necrotic enteritis occurs mostly in poorlymanaged herds living in poor hygienic conditions. It is frequently associated withclassic swine plague (cholera) or with such stress fac<strong>to</strong>rs as weaning <strong>and</strong> vaccination.The most frequent symp<strong>to</strong>ms are fever <strong>and</strong> diarrhea. The infection usually originatesfrom a carrier pig or contaminated food.Infection by other serotypes may sometimes give rise <strong>to</strong> serious outbreaks of salmonellosiswith high mortality.Because of the frequency with which swine are infected with different types ofsalmonellae, pork products have often been a source of human infection.SHEEP AND GOATS: Cases of clinical salmonellosis in these species are infrequent.The most <strong>common</strong> serotype found in gastroenteritis cases is S. typhimurium, butmany other serotypes have also been isolated. Serotype S. abortus ovis,which causesabortions in the last two months of pregnancy <strong>and</strong> gastroenteritis in sheep <strong>and</strong> goats,seems <strong>to</strong> be restricted <strong>to</strong> Europe <strong>and</strong> the Middle East (Timoney et al., 1988).HORSES: The most important pathogen among horses is S. abortus equi, whichcauses abortions in mares <strong>and</strong> arthritis in colts. It is distributed worldwide. As inother types of salmonellosis, predisposing fac<strong>to</strong>rs influence whether the infectionmanifests itself clinically. Pregnant mares are especially susceptible, particularly ifother debilitating conditions are present. Abortion occurs in the last months of pregnancy,<strong>and</strong> the fetus <strong>and</strong> placenta contain large numbers of bacteria. This serotype isadapted <strong>to</strong> horses <strong>and</strong> is rarely found in other animal species.Horses are also susceptible <strong>to</strong> other types of salmonellae, particularly S.typhimurium. Salmonella enteritis occurs in these animals, sometimes causing highmortality. Calves suffer from acute enteritis with diarrhea <strong>and</strong> fever; dehydrationmay be rapid. Nosocomial transmission has been seen in hospitalized horses. FromApril 1990 <strong>to</strong> January 1991, in an outbreak among hospitalized horses, 97.8% of theanimals contracted the infection due <strong>to</strong> S. typhimurium var. copenhagen with thesame plasmid profile. Other strains of S. typhimurium var. copenhagen with a differentplasmid profile <strong>and</strong> S. enteritidis began <strong>to</strong> appear in February 1991(Bauerfeind et al., 1992).


SALMONELLOSIS 239DOGS AND CATS: In recent years, a high prevalence of infection caused by numerousserotypes has been confirmed in cats <strong>and</strong> dogs. These animals may be asymp<strong>to</strong>maticcarriers or may suffer from gastroenteritic salmonellosis with varyingdegrees of severity.Dogs can contract the infection by eating the feces of other dogs, other domesticor peridomestic animals, or man. Dogs <strong>and</strong> cats can also be infected by contaminatedfood. In addition, dogs can transmit the disease <strong>to</strong> man.Treatment for these animals consists mainly of fluid <strong>and</strong> electrolyte replacement.Antibiotic treatment is reserved for septicemic cases <strong>and</strong> is effective if begun earlyin the course of the disease. The antibiotics indicated for invasive salmonellosis areampicillin, chloramphenicol, <strong>and</strong> sulfamethoxazole with trimethoprim (Timoney etal., 1988).Multiresistant animal strains that can be transmitted <strong>to</strong> man are another problem.The indiscriminate use of antibiotics in animals often results in changes in flora inthe colon, allowing rapid multiplication of resistant bacteria. In addition, the numberof carrier animals in the group that shed the etiologic agent can increase(Timoney et al., 1988).FOWL: Two serotypes, S. pullorum <strong>and</strong> S. gallinarum, are adapted <strong>to</strong> domesticfowl. They are not very pathogenic for man, although cases of salmonellosis causedby these serotypes have been described in children. Many other serotypes are frequentlyisolated from domestic poultry; for that reason, these animals are consideredone of the principal reservoirs of salmonellae.Pullorum disease, caused by serotype S. pullorum, <strong>and</strong> fowl typhoid, caused by S.gallinarum, produce serious economic losses on poultry farms if not adequatelycontrolled. Both <strong>diseases</strong> are distributed worldwide <strong>and</strong> give rise <strong>to</strong> outbreaks withhigh morbidity <strong>and</strong> mortality. Pullorum disease appears during the first 2 weeks oflife <strong>and</strong> causes high mortality. The agent is transmitted vertically as well as horizontally.Carrier birds lay infected eggs that contaminate incuba<strong>to</strong>rs <strong>and</strong> hatcheries.Fowl typhoid occurs mainly in adult birds <strong>and</strong> is transmitted by the fecal matter ofcarrier fowl. On an affected poultry farm, recuperating birds <strong>and</strong> apparently healthybirds are reservoirs of infection.Salmonellae unadapted <strong>to</strong> fowl also infect them frequently. In the US, more than200 serotypes of Salmonella spp. have been isolated from chickens <strong>and</strong>/or turkeys(Nagaraja et al., 1991). Nearly all the serotypes that attack man infect fowl as well.Some of these serotypes are isolated from healthy birds. The infection in adult birdsis generally asymp<strong>to</strong>matic, but during the first few weeks of life, its clinical pictureis similar <strong>to</strong> pullorum disease (loss of appetite, nervous symp<strong>to</strong>ms, <strong>and</strong> blockage ofthe cloaca with diarrheal fecal matter). The highest mortality occurs during the first2 weeks of life. Most losses occur between six <strong>and</strong> ten days after hatching.Mortality practically ceases after a month. The clinically apparent form of the diseaseis rare after three weeks of life, but many birds survive as carriers (Nagarajaet al., 1991).The most <strong>common</strong> agent in ducks <strong>and</strong> geese is S. typhimurium. The infection maybe transmitted from the infected ovary <strong>to</strong> the egg yolk, as in pullorum disease, or bycontamination of the shell when it passes through the cloaca.The most <strong>common</strong> agent of salmonellosis in pigeons is S. typhimurium var.copenhagen.


240 BACTERIOSESSalmonellosis is frequent in wild birds. In one species of seagull (Larus argentatus),it was found that 8.4% of 227 birds examined were carriers of salmonellae <strong>and</strong>that the serotypes were similar <strong>to</strong> those in man. Wild birds have also been implicatedas vec<strong>to</strong>rs of outbreaks of serotype S. montevideo in sheep <strong>and</strong> cattle in Scotl<strong>and</strong>(Butterfield et al., 1983; Coulson et al., 1983).OTHER ANIMALS: Rodents become infected with the serotypes prevalent in theenvironment in which they live. The rate of wild animal carriers is not high.Rodents found in <strong>and</strong> around food processing plants can be an important source ofhuman infection.Of 974 free-living wild animals examined in Panama, 3.4% were found <strong>to</strong> beinfected, principally by serotype S. enteritidis <strong>and</strong>, less frequently, by S. arizonae(Arizona hinshawii) <strong>and</strong> Edwardsiella. The highest rate of infection (11.8%) wasfound among the 195 marsupials examined. Salmonella was isolated from only 8 of704 spiny rats (Proechimys semispinosus).Outbreaks of salmonellosis among wild animals held in captivity in zoos or onpelt farms are not unusual.Salmonella infection in cold-blooded animals has merited special attention.Because of the high rate of infection among small turtles kept as house pets in theUS, their import was prohibited <strong>and</strong> a certificate stating them <strong>to</strong> be infection-freewas required for interstate commerce.An infection rate of 37% was found in 311 reptiles examined live or necropsiedat the National Zoo in Washing<strong>to</strong>n, D.C. The highest rate of infection was observedin snakes (55%) <strong>and</strong> the lowest in turtles (3%). The salmonellae isolated were 24different serotypes formerly classified under the <strong>common</strong> name of S. enteritidis, 1strain of S. choleraesuis, <strong>and</strong> 39 of S. arizonae. No disease in their hosts was attributed<strong>to</strong> these bacteria, but they may act <strong>to</strong>gether with other agents <strong>to</strong> cause opportunisticinfections (Cambre et al., 1980).Source of Infection <strong>and</strong> Mode of Transmission (Figure 17): Animals are thereservoir of zoonotic salmonellae. Practically any food of animal origin can be asource of infection for man. The most <strong>common</strong> vehicles are contaminated poultry,pork, beef, eggs, milk, <strong>and</strong> milk <strong>and</strong> egg products. Foods of vegetable origin contaminatedby animal products, human excreta, or dirty utensils, in both commercial processingplants <strong>and</strong> household kitchens, have occasionally been implicated as vehiclesof human salmonellosis. An outbreak of enteritis occurred in June <strong>and</strong> July of 1991 inthe US <strong>and</strong> Canada. It affected 400 people who ate melons contaminated by S. poona,a relatively rare serotype. It is assumed that the salmonellae penetrated the soft portionof the fruit from the rind when contaminated knives were used <strong>and</strong> the melons wereleft at ambient temperature in the summer (Publ Hlth News. Abst Hyg Comm Dis 60(9): 210, 1991). Contaminated public or private water supplies are important sourcesof infection in typhoid fever (S. typhi) <strong>and</strong>, less frequently, in other salmonella infections.An outbreak caused by S. typhimurium occurred in 1965 in Riverside, Californiadue <strong>to</strong> contaminated water. The causal agent was isolated from 100 patients examined,though it probably affected 16,000 people (Aserkoff et al., 1970).Fowl (chickens, turkeys, <strong>and</strong> ducks) represent the most important reservoir ofsalmonellae entering the human food chain (D’Aoust, 1989). In Engl<strong>and</strong> <strong>and</strong> Walesfrom 1981 <strong>to</strong> 1983, 51.3% of 347 vehicles of human salmonellosis were associatedwith fowl; in 1984–1985, 32.2% of 177 vehicles were of avian origin (Humphrey et


SALMONELLOSIS 241Figure 17. Salmonellosis. Mode of transmission (except Salmonellatyphi <strong>and</strong> the paratyphoid serotypes).Sick orcarrieranimalsof manyspeciesFecesContaminated food<strong>and</strong> environmentIngestionSusceptibleanimalsof manyspeciesIngestion of foodsof animal originWater <strong>and</strong> vegetablesManFecal-oral route(especially in hospitals)Manal., 1988). Another important source of salmonellae is raw or poorly cooked eggs,whether alone or as a component of various foods. An outbreak of S. enteritidis duringa wedding celebration in a London hotel affected 173 people. The agent, phagetype 4, was isolated from 118 of those affected <strong>and</strong> another 17 asymp<strong>to</strong>matic people.The source of infection was a sauce made from eggs imported from the continent.An unusual aspect of this outbreak was that some people fell ill only threehours after consuming the food. The percentage of eggs infected with S. enteritidisis low (an estimated 0.001%) but the risk increases when a large number of eggs isused <strong>to</strong> prepare a dish (Stevens et al., 1989). Pork, beef, milk, <strong>and</strong> milk products (icecream, cheeses) are other sources of human infection. Important contributing fac<strong>to</strong>rsare inadequate cooking, slow cooling of the food, lack of refrigeration for manyhours, <strong>and</strong> inadequate reheating before serving. Large outbreaks are invariably due<strong>to</strong> improper h<strong>and</strong>ling of food in restaurants <strong>and</strong> institutional dining facilities. Mancan also contract the infection directly from domestic animals or house pets, such asdogs, turtles, monkeys, hamsters, <strong>and</strong> others. Young children are especially susceptible<strong>to</strong> salmonellae in reptiles, even without direct contact. In Indiana (USA), twocases were described in children; one child was less than 2 weeks old <strong>and</strong> the otherless than 3 months. They were infected indirectly by S. marina. The source of theinfection was iguanas kept in the house. These animals harbor a wide variety ofserotypes <strong>and</strong> their rate of infection varies from 36% <strong>to</strong> 77% (CDC, 1992b). Thereare numerous reports from Asia, Canada, the US, <strong>and</strong> Europe on transmission fromsmall turtles <strong>to</strong> humans, particularly children. This led several countries <strong>to</strong> prohibittheir import. The serotypes isolated most frequently are S. poona <strong>and</strong> S. arizonae(D’Aoust et al., 1990). The long period of survival (many months) of salmonellae in


242 BACTERIOSESfecal matter can explain why direct contact is not always necessary, as in the case ofsome reptiles kept in or near a house (Morse <strong>and</strong> Duncan, 1974). Interhuman transmissionis particularly important in hospitals; children <strong>and</strong> the elderly are the principalvictims. In Baden-Würtenberg (Germany), an outbreak of S. enteritidis, phagetype 4, occurred in a home for elderly disabled persons. The same serotype <strong>and</strong>phage type was isolated from 95 residents <strong>and</strong> 14 employees. The source of infectionwas a dessert made of orange cream prepared with eggs with contaminatedshells (<strong>WHO</strong> Surveillance Programme for Control of Foodborne Infections <strong>and</strong>In<strong>to</strong>xications in Europe, 1991).Institutional <strong>and</strong> nosocomial outbreaks are usually due <strong>to</strong> food that is undercooked<strong>and</strong> kept at the wrong temperature, or <strong>to</strong> a kitchen employee who is anasymp<strong>to</strong>matic carrier. Nosocomial cases require prompt epidemiological investigationbecause they can involve patients who, given their age or illness, can experiencesevere cases of salmonellosis (CDC, 1991).Insects, particularly flies, may have some role as mechanical vec<strong>to</strong>rs in very contaminatedenvironments.Carrier animals perpetuate the animal-<strong>to</strong>-animal cycle by means of their excretaor, in the case of fowl, infected eggs. Feed contaminated by such ingredients asbone, meat, or fish meal plays an important role as a vehicle of infection.Intensive cattle-raising in developed countries is a very important contributingfac<strong>to</strong>r in the epidemiology of salmonellosis. Close contact between animals <strong>and</strong> theuse of concentrated feed or ingredients that may be contaminated create conditionsfavorable <strong>to</strong> outbreaks. In developing countries, the source of infection is mainly thecontaminated environment <strong>and</strong> water sources where animals crowd <strong>to</strong>gether.Animal-<strong>to</strong>-animal transmission occurs not only at the home establishment, butalso during shipping, at auctions <strong>and</strong> fairs, <strong>and</strong> even at slaughterhouses prior <strong>to</strong> sacrifice.Meat can become contaminated in abat<strong>to</strong>irs by means of contaminated equipment<strong>and</strong> utensils during skinning <strong>and</strong> butchering. Contaminated water can be asource of infection for man <strong>and</strong> animals.The cycle of infection in fowl begins with contaminated eggshells or yolks.Contaminated eggs spread the infection in the incuba<strong>to</strong>r. When the eggs hatch, thenewborn chicks become infected <strong>and</strong> many of those that do not die become carriers.This is the most important mechanism at work when fowl in poultry yards becomeinfected. Another vehicle of infection may be contaminated feed. Cannibalism <strong>and</strong>the ingestion of contaminated eggs also contribute <strong>to</strong> transmission of the infection.Non–species-specific serotypes spread easily from one animal species <strong>to</strong> another<strong>and</strong> also <strong>to</strong> humans.Role of Animals in the Epidemiology of the Disease: Since animals constitutethe reservoir of salmonellae (except S. typhi <strong>and</strong> the paratyphoid serotypes), theyplay an essential role in its epidemiology.Diagnosis: In humans, clinical diagnosis of gastroenteritis due <strong>to</strong> Salmonella isconfirmed by isolation of the etiologic agent from the patient’s s<strong>to</strong>ol, serologic typing,<strong>and</strong>, when necessary, phage typing <strong>and</strong> plasmid profiling. In the few cases ofsepticemia, the agent may be isolated from the blood during the first week of the disease,<strong>and</strong> from feces in the second <strong>and</strong> third weeks.Diagnosis of animal salmonellosis is also made by culturing fecal material. Forinfections caused by S. pullorum <strong>and</strong> S. gallinarum in fowl, serologic diagnosis is


SALMONELLOSIS 243important <strong>to</strong> identify <strong>and</strong> eliminate individual carriers. Infection by S. dublin can bediagnosed serologically in a herd, but not in individual cattle. As a screening test,the S. pullorum antigen can also be used in detecting antibodies for the lipopolysaccharideof S. enteritidis in chickens. Seroagglutination with the S. abortus equi antigencan be used as a preliminary test prior <strong>to</strong> culture in mares that have aborted.Postmortem examinations of animals primarily use cultures from the mesentericlymph nodes.Surveillance of food processing requires that cultures be made from product samplesat different stages of preparation, <strong>and</strong> from utensils <strong>and</strong> surfaces that come in<strong>to</strong>contact with the food. Special sampling methods have been developed for differentkinds of foods.Control: Given current conditions under which cattle <strong>and</strong> poultry are raised,transported, marketed, <strong>and</strong> slaughtered, as well as existing food processing practices,it is impossible <strong>to</strong> obtain salmonellae-free foods of animal origin. Control iscurrently based on protecting man from infection <strong>and</strong> reducing its prevalence in animals.Veterinary meat <strong>and</strong> poultry inspection <strong>and</strong> supervision of milk pasteurization<strong>and</strong> egg production are important for consumer protection.Another important control measure is the education of food h<strong>and</strong>lers, both incommercial establishments <strong>and</strong> in the home, about correct cooking <strong>and</strong> refrigerationpractices for foods of animal origin, <strong>and</strong> about personal <strong>and</strong> environmental hygiene.Epidemiological surveillance by health authorities is necessary <strong>to</strong> evaluate themagnitude of the problem in each country, locate the origins of outbreaks, <strong>and</strong> adoptmethods designed <strong>to</strong> reduce risks.In animals, salmonellosis control consists of: (a) elimination of carriers, whichis currently possible for pullorum disease <strong>and</strong> fowl typhoid by means of serologictests; (b) bacteriologic control of foods, mainly of such ingredients as fish,meat, <strong>and</strong> bone meal; (c) immunization; <strong>and</strong> (d) proper management of herds <strong>and</strong>poultry farms.Immunization may be an important method for preventing animal salmonellosis.Two types of vaccines are being used: bacterins <strong>and</strong> live attenuated vaccines.Bacterins are administered parenterally, usually in two doses two <strong>to</strong> four weeksapart. Commercially available bacterins act against S. dublin, S. typhimurium, <strong>and</strong>S. abortus equi. Live salmonellae vaccines are administered orally; they are usuallygenetically defective mutants. In the US, strains of S. dublin <strong>and</strong> S. typhimurium thatare unable <strong>to</strong> synthesize aromatic amino acids are used. Vaccines that are unable <strong>to</strong>synthesize purines are used against these serotypes <strong>and</strong> S. choleraesuis in Germany.These vaccines are avirulent <strong>and</strong> do not revert <strong>to</strong> the virulent state.In the US, a vaccine has been developed against S. choleraesuis, with a strainattenuated through repeated selection of a virulent strain with passes of neutrophilsfrom salmonellosis-free swine. In this way, the vaccine strain lost the 50-kilobaseplasmid, which is a virulence fac<strong>to</strong>r (Kramer et al., 1992). Live vaccines stimulatea greater cell-mediated immune response than bacterins, which primarily promote ahumoral response with little or no association with protection. Oral administration(whether of bacterins or live vaccines) has the advantage of producing local immunityin the intestine <strong>and</strong> reducing elimination of salmonellae in feces. Parenteraladministration of live vaccines can sometimes cause adverse reactions due <strong>to</strong> endo<strong>to</strong>xins(<strong>WHO</strong> Expert Committee on Salmonellosis Control, 1988).


244 BACTERIOSESThe results of many tests conducted <strong>to</strong> date indicate that immunization with vaccines<strong>and</strong> some bacterins can prevent the disease (particularly in its severe form), butnot the infection or carrier status.A control measure known as Nurmi’s method originated in Finl<strong>and</strong>. Salmonellafreecultures of fecal organisms from the cecum of adult birds are administeredorally <strong>to</strong> newly hatched chickens <strong>and</strong> turkey chicks. The cecal flora (some 60species of bacteria) from the adult birds compete with the salmonellae <strong>and</strong> thus protectthe chicks against salmonellosis at their most susceptible age. Treated chicksresist high doses of salmonellae. It is believed <strong>to</strong> work by competitive exclusion.Various countries have had success combating pullorum disease (S. pullorum) <strong>and</strong>fowl typhoid (S. gallinarum), reducing the rate of infection <strong>to</strong> a minimum. Severalcountries have undertaken control programs for S. enteritidis in fowl. Control isimportant <strong>to</strong> reduce both public health risk <strong>and</strong> economic losses. In general terms,the first step is <strong>to</strong> ensure that establishments that provide eggs for incubation <strong>and</strong> 1-day-old chicks are free of infection. Each group of egg layers must be examinedserologically <strong>and</strong> bacteriologically <strong>to</strong> certify those that are disease-free <strong>and</strong> destroythose that are infected. After the surroundings <strong>and</strong> the installations are disinfected,they should be repopulated from a safe source. Once a reliable source of eggs <strong>and</strong>chicks is assured, clean-up of commercial farms should begin.Some countries limit the control program <strong>to</strong> S. enteritidis, others <strong>to</strong> all invasiveserotypes, including S. typhimurium <strong>and</strong> S. hadar. In Sweden, during the first yearof control (1991–1992), infection was found in 6% of the layer establishments. Thisfell <strong>to</strong> 2% in the second year. An organization of broiler breeders in Northern Irel<strong>and</strong>had a successful program that eradicated infection by S. enteritidis in the establishmentsof their associates (McIlroy et al., 1989).BibliographyAger, E.A., F.H. Top, Sr. Salmonellosis. In:Top, F.H., Sr., P.F. Wehrle, eds. Communicable<strong>and</strong> Infectious Diseases. 7th ed. Saint Louis: Mosby; 1972.Aserkoff, B., S.A. Schroeder, P.S. Brachman. Salmonellosis in the United States—a fiveyearreview. Am J Epidemiol 92:13–24, 1970.Barker, R., D.C. Old, J.C. Sharp. Phage type/biotype groups of Salmonella typhimurium inScotl<strong>and</strong> 1974–6: Variation during spread of epidemic clones. J Hyg 84:115–125, 1980.Bauerfeind, R., L.H. Wieler, R. Weiss, G. Baljer. [Comparative plasmid profile analysis ofSalmonella typhimurium var. Copenhagen strains from a Salmonella outbreak in hospitalizedhorses.] Berl Münch Tierärztl Wochenschr 105:38–42, 1992.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Binsztein, N., T. Eiguer, M. D’Empaire. [An epidemic of salmonellosis in Buenos Aires <strong>and</strong>its suburbs.] Medicina 42:161–167, 1982.Bryan, F.L. Current trends in food-borne salmonellosis in the United States <strong>and</strong> Canada. JFood Protect 44:394–402, 1981.Butterfield, J., J.C. Coulson, S.V. Kearsey, P. Monaghan, J.H. McCoy, G.E. Spain. The herringgull Larus argentatus as a carrier of Salmonella. J Hyg 91:429–436, 1983.Bux<strong>to</strong>n, A., H.I. Field. Salmonellosis. In: Stableforth, A.W., I.A. Galloway, eds. InfectiousDiseases of Animals. London: Butterworth; 1959.


SALMONELLOSIS 245Cambre, R.C., D.E. Green, E.E. Smith, R.J. Montali, M. Bush. Salmonellosis <strong>and</strong> arizonosisin the reptile collection at the National Zoological Park. J Am Vet Med Assoc 177:800–803, 1980.Clarenburg, A. Salmonellosis. In: Van der Hoeden, J., ed. Zoonoses. Amsterdam: Elsevier;1964.Coulson, J.C., J. Butterfield, C. Thomas. The herring gull Larus argentatus as a likely transmittingagent of Salmonella montevideo <strong>to</strong> sheep <strong>and</strong> cattle. J Hyg 91:437–443, 1983.D’Aoust, J.Y. Salmonella. In: M.P. Doyle, ed. Foodborne Bacterial Pathogens. New York:Marcel Dekker; 1989.D’Aoust, J.Y., E. Daley, M. Crozier, A.M. Sewell. Pet turtles: A continuing internationalthreat <strong>to</strong> public health. Am J Epidemiol 132:233–238, 1990.Edwards, P.R., W.H. Ewing. Identification of Enterobacteriaceae. 3rd ed. Minneapolis:Burgess; 1972.Edwards, P.R., M.M. Gal<strong>to</strong>n. Salmonellosis. Adv Vet Sci 11:1–63, 1967.Eiguer, T., M.I. Caffar, G.B. Fronchkowsky. [Significance of Salmonella enteritidis in outbreaksof <strong>diseases</strong> transmitted by foods in Argentina, 1986–1988]. Rev Argent Microbiol22:31–36, 1990.Farmer, J.J., III, A.C. McWhorter, D.J. Brenner, et al. The Salmonella-Arizona group ofEnterobacteriaceae: Nomenclature, classification <strong>and</strong> reporting. Clin Microbiol Newsletter6:63–66, 1984.el-Gazzar, F.E., E.H. Marth. Salmonellae, salmonellosis, <strong>and</strong> dairy foods: A review. J DairySci 75:2327–2343, 1992.Gunn, R.A., Bullón, F. Salmonella enterocolitis: Report of a large foodborne outbreak inTrujillo, Peru. Bull Pan Am Health Organ 13(2):162–168, 1979.Hargrett-Bean, N.T., A.T. Pavia, R.V. Tauxe. Salmonella isolates from humans in the UnitedStates, 1984–1986. MMWR Morb Mortal Wkly Rep 37 (Suppl 2):25–31, 1988.Humphrey, T.J., G.C. Mead, B. Rowe. Poultry meat as a source of human salmonellosis inEngl<strong>and</strong> <strong>and</strong> Wales. Epidemiological Overview. Epidemiol Infect 100:175–184, 1988.Kourany, M., L. Bowdre, A. Herrer. Panamanian forest mammals as carriers of Salmonella.Am J Trop Med Hyg 25:449–455, 1976.Kramer, T.T., M.B. Roof, R.R. Matheson. Safety <strong>and</strong> efficacy of an attenuated strain ofSalmonella choleraesuis for vaccination of swine. Am J Vet Res 53:444–448, 1992.Le Minor, L., M.Y. Popoff. Request for an opinion. Designation of Salmonella enterica sp.nov., nom. rev., as the type <strong>and</strong> only species of the genus Salmonella. Int J Syst Bacteriol37:465–468, 1987.McIlroy, S.G., R.M. McCracken, S.D. Neill, J.J. O’Brien. Control, prevention <strong>and</strong> eradicationof Salmonella enteritidis infection in broiler <strong>and</strong> broiler breeder flocks. Vet Rec125(22):545–548, 1989.McLaren, I.M., C. Wray. Epidemiology of Salmonella typhimurium infection in calves:Persistence of salmonellae on calf units. Vet Rec 129:461–462, 1991.Morse, E.V., M.A. Duncan. Salmonellosis—an environmental health problem. J Am VetMed Assoc 165:1015–1019, 1974.Murray, M.J. Salmonella:Virulence fac<strong>to</strong>rs <strong>and</strong> enteric salmonellosis. J Am Vet Med Assoc189:145–147, 1986.Nagaraja, K.V., B.S. Pomeroy, J.E. Williams. Paratyphoid infections. In: Calnek, B.W., H.J.Barnes, C.W. Beard, W.M. Reid, H.W. Yoder, Jr., eds. Diseases of Poultry. 9th ed. Ames: IowaState University Press; 1991.Peluffo, C.A. Salmonellosis in South America. In:Van Ove, E., ed. The World Problem ofSalmonellosis. The Hague: Junk; 1964.Pietzsch, O. Salmonellose-Überwachtung in der Bundesrepublik Deutschl<strong>and</strong> einschl.Berlin (West). Bundesgesundhbl 25:325–327, 1982.Pietzsch, O. Salmonellose-Überwachtung bei Tieren, Lebens-und Futtermitteln in derBundesrepublik Deutschl<strong>and</strong> einschl. Berlin (West), 1984/85. Bundesgesundhbl 29:427–429, 1986.


246 BACTERIOSESPoehn, H.P. Salmonellose-Überwachtung beim Menschen in der BundesrepublikDeutschl<strong>and</strong> einschl. Berlin (West). Bundesgesundhbl 25:320–324, 1982.Rodrigue, D.C., R.V. Tauxe, B. Rowe. International increase in Salmonella enteritidis: Anew p<strong>and</strong>emic? Epidemiol Infect 105:21–27, 1990.Silliker, J.H. The Salmonella problem: Current status <strong>and</strong> future direction. J Food Protect45:661–666, 1982.Skerman, V.B.D., V. McGowan, P.H.A. Sneath. Approved list of bacterial names. Int J SystBacteriol 30:225–420, 1980.Smith, B.P., M. Reina-Guerra, S.K. Hoiseth, B.A. S<strong>to</strong>cker, F. Habasche, E. Johnson, et al.Aromatic-dependent Salmonella typhimurium as modified live vaccines for calves. Am J VetRes 45:59–66, 1984.Stevens, A., C. Joseph, J. Bruce, et al. A large outbreak of Salmonella enteritidis phage type4 associated with eggs from overseas. Epidemiol Infect 103:425–433, 1989.Tauxe, R.V., M.P. Tormey, L. Mascola, et al. Salmonellosis outbreak on transatlanticflights; foodborne illness on aircraft: 1947–1984. Am J Epidemiol 125:150–157, 1987.Taylor, J., J.H. McCoy. Salmonella <strong>and</strong> Arizona infections. In: Riemann, H., ed. Food-Borne Infections <strong>and</strong> In<strong>to</strong>xications. New York: Academic Press; 1969.Timoney, J.F., J.H. Gillespie, F.W. Scott, J.E. Barlough. Hagan <strong>and</strong> Bruner’s Microbiology<strong>and</strong> Infectious Diseases of Domestic Animals. 8th ed. Ithaca, New York: Coms<strong>to</strong>ck; 1988.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Salmonella Surveillance. Annual Summary 1980. Atlanta:CDC; 1982.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Update: Salmonella enteritidis infections <strong>and</strong> shell eggs—United States, 1990. MMWR Morb Mortal Wkly Rep 39:909–912, 1990.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Foodborne nosocomial outbreak of Salmonella reading—Connecticut. MMWR Morb Mortal Wkly Rep 40 (46):804–806, 1991.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Outbreak of Salmonella enteritidis infection associated withconsumption of raw shell eggs, 1991. MMWR Morb Mortal Wkly Rep 41:369–372, 1992a.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Iguana-associated salmonellosis—Indiana, 1990. MMWRMorb Mortal Wkly Rep 41 (3):38–39, 1992b.United States of America, National Academy of Sciences, National Research Council. Anevaluation of the Salmonella problem. Washing<strong>to</strong>n, D.C.: National Academy of Sciences; 1969.<strong>WHO</strong> Expert Committee on Microbiological Aspects of Food Hygiene. Microbiologicalaspects of food hygiene. Report of a <strong>WHO</strong> expert committee with the participation of FAO.Geneva: <strong>WHO</strong>; 1968. (Technical Report Series 399).<strong>WHO</strong> Expert Committee on Salmonellosis Control. Salmonellosis Control: The Role ofAnimal <strong>and</strong> Product Hygiene. Report of a <strong>WHO</strong> Expert Committee. Geneva: <strong>WHO</strong>, 1988.(Technical Report Series 774).<strong>WHO</strong> Scientific Working Group. Enteric infections due <strong>to</strong> Campylobacter, Yersinia,Salmonella, <strong>and</strong> Shigella. Bull World Health Organ 58:519–537, 1980.<strong>WHO</strong> Surveillance Programme for Control of Foodborne Infections <strong>and</strong> In<strong>to</strong>xications inEurope N° 28:4–5, 1991.Wilkins, E.G., C. Roberts. Extraintestinal salmonellosis. Epidemiol Infect 100:361–368,1988.Williams, L.P., B.C. Hobbs. Enterobacteriaceae infections. In: Hubbert, W.T., W.P.McCulloch, P.R. Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed.Springfield: Thomas; 1975.


SHIGELLOSIS 247SHIGELLOSISICD-10 A03.0 shigellosis due <strong>to</strong> Shigella dysenteriae;A03.1 shigellosis due <strong>to</strong> Shigella flexneri;A03.2 shigellosis due <strong>to</strong> Shigella boydii;A03.3 shigellosis due <strong>to</strong> Shigella sonnei; A03.8 other shigellosisSynonyms: Bacillary dysentery.Etiology: The genus Shigella belongs <strong>to</strong> the family Enterobacteriaceae. Shigellaeare small gram-negative, nonmotile, unencapsulated bacilli; they are anaerogenic(with a few exceptions) <strong>and</strong> non-lac<strong>to</strong>se fermenting (or slow fermenters).The genus Shigella may be considered genetically as a single species, closelyrelated in DNA analyses <strong>to</strong> E. coli. However, it is divided in<strong>to</strong> four species based onphenotype traits. Each species is a distinct serogroup: Shigella dysenteriae(serogroup A), S. flexneri (serogroup B), S. boydii (serogroup C), <strong>and</strong> S. sonnei(serogroup D). The four serogroups contain a <strong>to</strong>tal of 38 serotypes. Serotyping isimportant in epidemiological investigation. Diagnostic labora<strong>to</strong>ries are generallylimited <strong>to</strong> identifying the serogroup <strong>and</strong> sending cultures <strong>to</strong> a reference labora<strong>to</strong>ryfor identification of the serotype.The primary virulence fac<strong>to</strong>r of a Shigella strain is its ability <strong>to</strong> invade cells of theintestinal mucosa. The invasive capacity depends on fac<strong>to</strong>rs controlled by both chromosomic<strong>and</strong> plasmidic genes (Keusch <strong>and</strong> Bennish, 1991).The invasive capacity of a strain can be demonstrated with the Sereny test, whichconsists of inoculating a culture in a guinea pig’s conjunctival sac. Invasive strainsproduce kera<strong>to</strong>conjunctivitis in 24 <strong>to</strong> 48 hours. Cultures obtained from clinical casesalways yield a positive Sereny test result. Shigellae also produce cy<strong>to</strong><strong>to</strong>xins, particularlyin the case of S. dysenteriae serotype 1 (Shiga <strong>to</strong>xin).Geographic Distribution: Worldwide.Occurrence in Man: Shigellosis can be either epidemic or endemic. Epidemic orp<strong>and</strong>emic shigellosis is usually caused by S. dysenteriae serotype 1 (Shiga bacillus),the most virulent <strong>and</strong> <strong>to</strong>xigenic strain. In 1969–1970, a widespread epidemic causedby S. dysenteriae serotype 1 occurred in Central America <strong>and</strong> Mexico, with highmorbidity <strong>and</strong> mortality rates, particularly in children. More than 13,000 patientsdied as a result. The infection was introduced in the US, where 140 cases occurredfrom 1970 <strong>to</strong> 1972. The epidemic spread <strong>to</strong> Central Africa <strong>and</strong> Asia (India,Bangladesh, <strong>and</strong> Sri Lanka). Plasmid analysis demonstrated that the p<strong>and</strong>emic wasnot produced by a single bacterial clone. Thus, it is difficult <strong>to</strong> explain the appearanceof the disease in such distant areas. The strains isolated in all areas proved <strong>to</strong>be multiresistant <strong>to</strong> antibiotics (<strong>WHO</strong>, 1987). In Guatemala, there were 112,000cases with 10,000 deaths from 1969 <strong>to</strong> 1972. A new outbreak appeared in Guatemalain 1991 caused by S. dysenteriae serotype 1; it affected 540 people in the course ofone month, both in Guatemala City <strong>and</strong> in Verapaz, a city of 10,000 inhabitants(CDC, 1991).Endemic shigellosis is usually caused by S. flexneri <strong>and</strong> S. sonnei. The first occursprimarily in developing countries, the second, in economically advanced countries.Morbidity <strong>and</strong> mortality rates are high in developing countries, particularly in chil-


248 BACTERIOSESdren aged 1 <strong>to</strong> 5 years (<strong>WHO</strong>, 1987). Of 16,567 isolations done in the US during1987, 67.7% were S. sonnei, 22.2% were S. flexneri, 2.1% were S. boydii, 1.4% wereS. dysenteriae, <strong>and</strong> 6.6% were unidentified species (Keusch <strong>and</strong> Bennish, 1991).It is very difficult <strong>to</strong> calculate the number of cases worldwide, but they are estimatedat more than 200 million each year, 650,000 of whom die (<strong>WHO</strong>, 1991). Inthe US, there are an estimated 300,000 clinical cases (Bennett, cited in Wachsmuth<strong>and</strong> Morris, 1989).An outbreak affecting large numbers of people occurred in 1987 during a mass“Rainbow Family” gathering in a forest in North Carolina (USA). It is estimatedthat more than 50% of the 12,700 participants were affected. The location’s sanitaryinfrastructure was insufficient for so many people. The outbreak was causedby S. sonnei, which is resistant <strong>to</strong> many antibiotics (ampicillin, tetracycline, <strong>and</strong>trimethoprim with sulfamethoxazole), <strong>and</strong> which contained a 90-kilobase plasmidnot found in strains not related <strong>to</strong> this epidemic. When the participants dispersed,they became the source of infection for outbreaks in three US states (Whar<strong>to</strong>n etal., 1990).Those who suffer most from the disease are those who cannot follow personalhygiene rules, such as patients or residents confined in different institutions.Children are the principal victims of the disease in endemic areas. Resistance inadults is due <strong>to</strong> acquired immunity <strong>to</strong> the prevalent serotype. Adult travelers visitingendemic areas contract the disease because they have had no previous exposure.Similarly, when a new serotype is introduced in<strong>to</strong> a susceptible population, the diseaseaffects all age groups (Levine <strong>and</strong> Lanata, 1983).Occurrence in Animals: It is <strong>common</strong> in captive nonhuman primates <strong>and</strong> rare inother animal species. All species of Shigella, including S. dysenteriae type 1 (Shigabacillus), which is considered the most pathogenic form for man, have been isolatedfrom nonhuman primates (L’Hote, 1980). In 1984, an epizootic caused by S. flexnerioccurred at the National Zoo in Washing<strong>to</strong>n, DC (USA), <strong>and</strong> since then, shigellosishas become endemic. The infected species were gibbons (Hylobates concolor <strong>and</strong>H. syndactylies), macaques (Macaca silenus, M. nigra, <strong>and</strong> M. sylvanus), colobusmonkeys (Colobus guerzea), <strong>and</strong> gorillas (Gorilla gorilla). From 1984 <strong>to</strong> 1988, thetwo species of gibbons (species in danger of extinction) had a high rate of infection<strong>and</strong> disease (Banish et al., 1993a). S. sonnei was isolated from mangabey monkeys(Cercocebus albigena) <strong>and</strong> spider monkeys (Ateles susciceps) in the same zoologicalcollection (Banish et al., 1993a).The Disease in Man: It is seen most often in preschool-aged children. When anew serotype is introduced in tropical areas in which the population is undernourished,the disease affects all age groups, particularly children, the elderly, <strong>and</strong> debilitatedindividuals. The incubation period is one <strong>to</strong> seven days, but usually four days.The clinical picture may vary from an asymp<strong>to</strong>matic infection <strong>to</strong> a serious <strong>and</strong>fatal disease. The disease begins with fever <strong>and</strong> abdominal pains, as well as diarrheathat may be watery at first <strong>and</strong> later dysenteric with blood <strong>and</strong> mucus. The rectum<strong>and</strong> colon are the parts of the intestine most affected. In the final stages, there is anintense tenesmus with frequent elimination of small amounts of feces consistingalmost entirely of blood <strong>and</strong> mucus. The disease is self-limiting in well-nourishedindividuals, but may last for weeks or months in undernourished persons (Keusch<strong>and</strong> Bennish, 1991). Convulsions are frequent in hospitalized children.


SHIGELLOSIS 249Shigellae rapidly acquire resistance <strong>to</strong> antimicrobials. The choice of an antimicrobialwill depend on the antibiogram of the strain isolated or local patterns of susceptibility.Fluids <strong>and</strong> electrolytes must be replaced if dehydration occurs.Antiperistaltics are contraindicated, both for intestinal infections caused by shigellae<strong>and</strong> for other intestinal infections.In many countries, strains of Shigella resistant <strong>to</strong> sulfonamides <strong>and</strong> <strong>to</strong> severalantibiotics have been observed.The Disease in Animals: It occurs in monkeys, with a clinical picture similar <strong>to</strong>that in man. In nonhuman primate colonies, strains resistant <strong>to</strong> many antibiotics arefrequently found. As in man, an antibiogram must be done <strong>to</strong> identify the mostappropriate antimicrobial. Enrofloxacine was used with good results at the NationalZoo in Washing<strong>to</strong>n, DC (Banish et al., 1993a).Source of Infection <strong>and</strong> Mode of Transmission: The principal reservoir of theinfection for man is other humans who are sick or carriers. The sources of infectionare feces <strong>and</strong> contaminated objects. The most <strong>common</strong> mode of transmission is thefecal-oral route. Outbreaks with numerous cases have had their origin in a <strong>common</strong>source of infection, such as foods contaminated by h<strong>and</strong>s or feces of carrier individuals.Insects, particularly flies, can also play a role as mechanical vec<strong>to</strong>rs.There is a direct relationship between the frequency of shigellosis <strong>and</strong> a country’sdegree of economic development, as well as between poor <strong>and</strong> well-off classeswithin a country. Lack of health education, health infrastructure (potable water <strong>and</strong>sewer system), environmental hygiene, <strong>and</strong> personal hygiene habits are all fac<strong>to</strong>rsthat contribute <strong>to</strong> the spread of infection. Shigellosis is a disease of poverty.Bacillary dysentery is a serious disease with high mortality in nonhuman primatesin captivity, but there is doubt that monkeys can harbor the etiologic agent in theirnatural habitat. Monkeys probably contract the infection though contact withinfected humans. The infection spreads rapidly in nonhuman primate coloniesbecause the monkeys defecate on the cage floor <strong>and</strong> often throw their food there.Role of Animals in the Epidemiology of the Disease: Of little significance.Cases of human bacillary dysentery contracted from nonhuman primates are known.The victims are mainly children. In highly endemic areas, dogs may shed Shigella,at least temporarily.The etiologic agent has been isolated rarely from bats <strong>and</strong> rattlesnakes.Nevertheless, animals other than nonhuman primates play an insignificant role.Diagnosis: Definitive diagnosis depends on isolation of the etiologic agent by cultureof fecal material on selective media. There are several selective media, which arebased on the suppression of lac<strong>to</strong>se fermenters. One of these is MacConkey agar withbile salts, xylose, lysine, <strong>and</strong> deoxycholate (XLD). Serologic identification <strong>and</strong> typing,at least of the serogroup, are important for diagnosis <strong>and</strong> for epidemiological research.Control: In man, control measures include: (a) environmental hygiene, especiallydisposal of human waste <strong>and</strong> provision of potable water; (b) personal hygiene; (c)education of the public <strong>and</strong> of food h<strong>and</strong>lers about the sources of infection <strong>and</strong>modes of transmission; (d) sanitary supervision of the production, preparation, <strong>and</strong>preservation of foods; (e) control of flies; (f) reporting <strong>and</strong> isolation of cases <strong>and</strong>sanitary disposal of feces; <strong>and</strong> (g) search for contacts <strong>and</strong> the source of infection.


250 BACTERIOSESA live strep<strong>to</strong>mycin-dependent vaccine, administered orally in three or four doses,provided good protection against the clinical disease for 6 <strong>to</strong> 12 months. However,it is currently not in use due <strong>to</strong> side effects such as vomiting in a small number ofthose who have been given the vaccine. Another undesirable <strong>and</strong> more serious effec<strong>to</strong>f this vaccine is the instability of the strain <strong>and</strong> reversion <strong>to</strong> its original virulence(<strong>WHO</strong>, 1991). Different types of vaccines have been developed, including hybridsof Shigella <strong>and</strong> E. coli, <strong>and</strong> of Shigella <strong>and</strong> Salmonella; vaccines obtained throughdeletions <strong>and</strong> mutations; <strong>and</strong> oral vaccines of dead shigellae. All these vaccines areawaiting evaluation (<strong>WHO</strong>, 1991).In two military camps in Israel, intensive measures (primarily bait <strong>and</strong> strategicallylocated traps) were taken in the early summer of 1988 <strong>to</strong> control flies for 11weeks. The test was repeated in the summer of 1989. The number of flies wasreduced by 64% <strong>and</strong> clinic visits for diarrhea caused by shigellosis fell by 42% inthe first year <strong>and</strong> 85% in the second. These results indicate that flies, acting asmechanical vec<strong>to</strong>rs, are an important fac<strong>to</strong>r in the transmission of shigellosis (Cohenet al., 1991).Indiscriminate use of antibiotics must be avoided in order <strong>to</strong> prevent the emergenceof multiresistant strains <strong>and</strong> <strong>to</strong> ensure that these medications remain availablefor use in severe cases.In animals, control consists of: (a) isolation <strong>and</strong> treatment of sick or carrier monkeys;(b) careful cleaning <strong>and</strong> sterilization of cages; (c) prevention of crowding incages; <strong>and</strong> (d) prompt disposal of wastes <strong>and</strong> control of insects.At the National Zoo in Washing<strong>to</strong>n, DC, carrier status for multiresistant S. flexneriwas eliminated through intramuscular administration of enrofloxacine (5 mg/kg ofbodyweight every 24 hours for 10 days). The large primates received the same medicineorally. In this way, it was possible <strong>to</strong> eradicate S. flexneri from a colony of 85primates, although after 10 <strong>to</strong> 12 months S. sonnei was isolated from the feces ofthree animals (Banish et al., 1993b).BibliographyBanish, L.D., R. Sims, D. Sack, et al. Prevalence of shigellosis <strong>and</strong> other enteric pathogensin a zoologic collection of primates. J Am Vet Med Assoc 203:126–132, 1993a.Banish, L.D., R. Sims, M. Bush, et al. Clearance of Shigella flexneri carriers in a zoologiccollection of primates. J Am Vet Med Assoc 203:133–136, 1993b.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bennett, J.V. Cited in: Wachsmuth, K., G.K. Morris. Shigella. In: Doyle, M.P. FoodborneBacterial Pathogens. New York: Marcel Dekker; 1989.Cohen, D., M. Green, M. Block, et al. Reduction of transmission of shigellosis by controlof houseflies (Musca domestica). Lancet 337:993–997, 1991.Edwards, P.R., W.H. Ewing. Identification of Enterobacteriaceae. 3rd ed. Minneapolis:Burgess; 1972.Fiennes, R. Zoonoses of Primates. Ithaca: Cornell University Press; 1967.Keusch, G.T., M.L. Bennish. Shigellosis. In: Evans, A.S., P.S. Brachman, eds. BacterialInfections of Humans. 2nd ed. New York: Plenum Medical Book Co.; 1991.Keusch, G.T., S.B. Formal. Shigellosis. In: Warren, K.S., A.A.F. Mahmoud, eds. Tropical<strong>and</strong> Geographical Medicine. New Jersey: McGraw-Hill; 1984.


STAPHYLOCOCCAL FOOD POISONING 251Levine, M.M., C. Lanata. Progresos en vacunas contra diarrea bacteriana. Adel MicrobiolEnf Inf 2:67–118, 1983.Lewis, J.N., E.J. Gangarosa. Shigellosis. In:Top, F.H., Sr., P.F. Wehrle, eds. Communicable<strong>and</strong> Infectious Diseases. 7th ed. Saint Louis: Mosby; 1972.L’Hote, J.L. Contribution à l’étude des salmonelloses et des shigelloses des primates.Zoonoses [thesis]. École Nationale Vétérinaire de Lyon, 1980.Ruch, T.C. Diseases of Labora<strong>to</strong>ry Primates. Philadelphia: Saunders; 1959.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Shigella dysenteriae type 1—Guatemala, 1991. MMWR MorbMortal Wkly Rep 40(25):421, 427–428, 1991.Wachsmuth, K., G.K. Morris. Shigella. In: Doyle, M.P. Foodborne Bacterial Pathogens.New York: Marcel Dekker; 1989.Whar<strong>to</strong>n, M., R.A. Spiegel, J.M. Horan, et al. A large outbreak of antibiotic-resistantshigellosis at a mass gathering. J Infect Dis 162:1324–1328, 1990.<strong>WHO</strong> Scientific Working Group. Enteric infections due <strong>to</strong> Campylobacter, Yersinia,Salmonella, <strong>and</strong> Shigella. Bull World Health Organ 58:519–537, 1980.Williams, L.P., B.C. Hobbs. Enterobacteriaceae infections. In: Hubbert, W.T., W.F.McCulloch, P.R. Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed.Springfield: Thomas; 1975.World Health Organization (<strong>WHO</strong>). Development of vaccines against shigellosis:Memor<strong>and</strong>um from a <strong>WHO</strong> meeting. Bull World Health Organ 65:17–25, 1987.World Health Organization (<strong>WHO</strong>). Research priorities for diarrhoeal disease vaccines:Memor<strong>and</strong>um from a <strong>WHO</strong> meeting. Bull World Health Organ 69(6):667–676, 1991.STAPHYLOCOCCAL FOOD POISONINGICD-10 A05.0 foodborne staphylococcal in<strong>to</strong>xicationSynonyms: Staphylococcal alimentary <strong>to</strong>xicosis, staphylococcal gastroenteritis.Etiology: It is caused by an entero<strong>to</strong>xin preformed in food by Staphylococcusaureus. The overwhelming majority of outbreaks are due <strong>to</strong> coagulase-positivestrains of S. aureus. Very few coagulase-negative strains are capable of producingentero<strong>to</strong>xins. Some outbreaks may be due <strong>to</strong> S. intermedius <strong>and</strong> S. hyicus.The genus Staphylococcus consists of gram-positive bacteria in the form of coccigrouped in clusters. The bacteria is not very heat-resistant, but the entero<strong>to</strong>xin is.There are five known types of entero<strong>to</strong>xins (A, B, C, D, <strong>and</strong> E), but entero<strong>to</strong>xin A ismost prevalent in outbreaks. Some strains of S. aureus can produce both the entero<strong>to</strong>xins<strong>and</strong> <strong>to</strong>xic shock syndrome <strong>to</strong>xin-1.Geographical Distribution: Worldwide.Occurrence in Man: In some countries, the disease is an important cause of foodpoisoning. Most sporadic cases are not recorded. Outbreaks affecting several ormany people are those that are primarily known <strong>and</strong> recorded.


252 BACTERIOSESIn the US during the period 1977–1981, 131 outbreaks were reported, affecting7,126 people. In the last three years of that five-year period, only entero<strong>to</strong>xin A wasincriminated. Milk (the most <strong>common</strong> source of <strong>to</strong>xins C <strong>and</strong> D) <strong>and</strong> commerciallypackaged foods are the least <strong>common</strong> causes of the disease in the United States(Holmberg <strong>and</strong> Blake, 1984). In Japan, the annual average of food poisoning outbreaksfrom 1976 <strong>to</strong> 1980 was 827. Of a <strong>to</strong>tal of 8,742 cases, 28.2% were caused bystaphylococcal poisoning (Genigeorgis, 1989).It has been suggested that a proportion of the intestinal disorders frequentlyobserved in developing countries are caused by staphylococcal food poisoning.Evidence of this is the fact that titers of antibodies <strong>to</strong> entero<strong>to</strong>xins are higher in residentsof these countries than in travelers (Bergdoll, 1979).Occurrence in Animals: Spontaneous cases of staphylococcal poisoning indomestic animals are not known. The rhesus monkey (Macaca mulatta) is susceptible<strong>to</strong> the entero<strong>to</strong>xin through the digestive tract <strong>and</strong> is used as an experimental animal<strong>to</strong> show the presence of the <strong>to</strong>xin in implicated foods. Intravenous or peri<strong>to</strong>nealinoculation with the entero<strong>to</strong>xin in cats <strong>and</strong> kittens has also been used for the samepurpose. Dogs possibly suffer from gastroenteritis similar <strong>to</strong> that in man.Mastitis in cattle caused by staphylococci is of interest from a public health perspective.In modern milking systems, S. aureus is a <strong>common</strong> pathogen in cows’udders. The agent is transmitted by means of milking machines or the milker’sh<strong>and</strong>s, <strong>and</strong> enters through the milk duct or superficial lesions on the teat. Mastitiscaused by S. aureus in cattle may vary from the prevalent subclinical form of infection<strong>to</strong> a severe gangrenous form. Both forms are economically important becauseof the losses they cause in milk production (Gillespie <strong>and</strong> Timoney, 1981). Studiesconducted in five northern European countries <strong>and</strong> in Japan have shown that a largeproportion of the staphylococci isolated from cases of bovine mastitis are <strong>to</strong>xigenic.In Europe, 41.4% of 174 strains isolated produced entero<strong>to</strong>xins, <strong>and</strong> of these, 48.6%produced A; 5.6%, B; 29.2%, C; <strong>and</strong> 33.3%, D; either singly or in combination. InJapan, 34.4% of 1,056 strains isolated from cows with subclinical mastitis were <strong>to</strong>xigenic,<strong>and</strong> of these 31.1% produced entero<strong>to</strong>xin A; 54.3%, C; 27%, D; <strong>and</strong> 10.7%,B; either singly or in combination. Entero<strong>to</strong>xins A, C, or D are the predominanttypes in staphylococcal poisoning in many countries (Ka<strong>to</strong> <strong>and</strong> Kume, 1980).Nevertheless, the types of entero<strong>to</strong>xins produced by strains isolated from milk seem<strong>to</strong> vary in prevalence in different countries; this may often be because an unrepresentativenumber of strains has been studied.S. intermedius <strong>and</strong> S. aureus are the most <strong>common</strong> agents in canine skin infections<strong>and</strong> cause pyoderma, impetigo, folliculitis, <strong>and</strong> furunculosis. S. aureus is frequentlya complicating agent of demodectic mange, producing cellulitis in the deeplayers of the skin. Entero<strong>to</strong>xigenic staphylococci were isolated from 13% of 115domestic dogs in Japan. The strains isolated were producers of entero<strong>to</strong>xins A, C,<strong>and</strong> D that can cause food poisoning in man (Kaji <strong>and</strong> Ka<strong>to</strong>, 1980). A study conductedin Brazil in dogs with pyodermatitis confirmed that 13 of 52 isolates of S.intermedius <strong>and</strong> 6 of 21 of S. aureus produced entero<strong>to</strong>xins. There were six isolatesof entero<strong>to</strong>xin C, seven of D, <strong>and</strong> six of E. Four strains produced <strong>to</strong>xic shock syndrome<strong>to</strong>xin-1 (Hirooka et al., 1988).In fowl, staphylococcal infection can cause <strong>diseases</strong> ranging from pyoderma <strong>to</strong>septicemia with different localizations (salpingitis, arthritis, <strong>and</strong> other disorders).


STAPHYLOCOCCAL FOOD POISONING 253Purulent staphylococcal synovitis is a disease that causes appreciable losses inchickens <strong>and</strong> turkeys. In the former Czechoslovakia, one of the principal sources ofstaphylococcal food poisoning is thought <strong>to</strong> be infected poultry (Raska et al., 1980).Staphylococcal strains isolated from poultry farms in that country <strong>and</strong> others produceentero<strong>to</strong>xin D. Many researchers have isolated S. aureus from the nasal passages<strong>and</strong> skin of 100% of the birds examined, as well as from the nose <strong>and</strong> skin of72% of swine (Genigeorgis, 1989). These data indicate that meat- <strong>and</strong> milk-producinganimals may make a significant contribution <strong>to</strong> contamination of the food chain.The Disease in Man: The incubation period is short, generally three hours afteringestion of the food involved. The interval between ingestion <strong>and</strong> the first symp<strong>to</strong>msmay vary from 30 minutes <strong>to</strong> 8 hours depending on the amount of <strong>to</strong>xiningested <strong>and</strong> the susceptibility of the individual.The major symp<strong>to</strong>ms are nausea, vomiting, abdominal pain, <strong>and</strong> diarrhea. Somepatients may show low fever (up <strong>to</strong> 38°C). More serious cases may also show prostration,cephalalgia, abnormal temperature, <strong>and</strong> lowered blood pressure as well asblood <strong>and</strong> mucus in the s<strong>to</strong>ol <strong>and</strong> vomit. The course of the disease is usually benign<strong>and</strong> the patient recovers without medication in 24 <strong>to</strong> 72 hours.There are patients who require hospitalization due <strong>to</strong> the severity of the symp<strong>to</strong>ms.It is assumed that these are people who have ingested foods with high dosesof the entero<strong>to</strong>xin, who were not exposed <strong>to</strong> the entero<strong>to</strong>xin in the past, or who maybe debilitated due <strong>to</strong> other causes.Source of Infection <strong>and</strong> Mode of Transmission: The principal reservoir of S.aureus is the human carrier. A high proportion of healthy people (30% <strong>to</strong> 35%) havestaphylococci in the nasopharynx <strong>and</strong> on the skin. A carrier with a respira<strong>to</strong>ry diseasecan contaminate foods by sneezing, coughing, or expec<strong>to</strong>rating. Similarly, hemay contaminate food he h<strong>and</strong>les if he has a staphylococcal skin lesion. However,even if not sick himself, the carrier may contaminate food by h<strong>and</strong>ling different foodingredients, equipment, utensils, or the finished product. According <strong>to</strong> variousauthors, the proportion of entero<strong>to</strong>xin-producing S. aureus strains of human originvaries from 18% <strong>to</strong> 75% (Pulverer, 1983). The proportion of <strong>to</strong>xigenic strains isolatedfrom various sources (humans, animals, <strong>and</strong> food) is very high.Strains of human origin predominate in epidemics, but animals are also reservoirsof the infection. Milk from cow udders infected with staphylococci can contaminatenumerous milk products. Many outbreaks of staphylococcal poisoning have beencaused by the consumption of inadequately refrigerated raw milk or cheeses fromcows whose udders harbored staphylococci. The largest outbreak affected at least500 students in California (USA) between 1977 <strong>and</strong> 1981 <strong>and</strong> was traced <strong>to</strong> chocolatemilk (Holmberg <strong>and</strong> Blake, 1984). Another outbreak occurred in the US inwhich 850 students became ill after drinking chocolate milk. The average amount ofentero<strong>to</strong>xin A in the milk was 144 ng per half-pint car<strong>to</strong>n (Evenson et al., 1988).Goat milk is implicated more rarely. A small outbreak occurred in Israel amongBedouin children who drank semna, goat milk that is skimmed, sweetened, <strong>and</strong>heated. The milk came from a goat with mastitis caused by S. aureus entero<strong>to</strong>xin B(Gross et al., 1988).Small outbreaks <strong>and</strong> sporadic cases occurred in a <strong>to</strong>wn in Scotl<strong>and</strong> betweenDecember 1984 <strong>and</strong> January 1985, in which cheeses made from sheep milk wereimplicated. Bacteriological examination of the various samples of the cheese was


254 BACTERIOSESunsuccessful in isolating Staphylococcus, but the presence of entero<strong>to</strong>xin A wasconfirmed (Bone et al., 1989). In various Mediterranean countries, Staphylococcusis one of the most important agents in ovine mastitis. Not only can ovine staphylococcuscause economic losses, it could also be a public health problem. Food poisoningis probably the most important foodborne disease in Spain <strong>and</strong> other countriesof the region. The vehicle of poisoning could be cheese made from sheep’smilk. In Spain, 46 of 59 isolates of S. aureus produced entero<strong>to</strong>xin C; 2, entero<strong>to</strong>xinA; 1, entero<strong>to</strong>xin D; <strong>and</strong> 2, entero<strong>to</strong>xins A <strong>and</strong> C (Gutiérrez et al., 1982). In developingcountries, where the refrigeration of milk after milking leaves much <strong>to</strong> bedesired, it is possible that milk <strong>and</strong> milk products are an important source of staphylococcalin<strong>to</strong>xication.According <strong>to</strong> recent studies, a high proportion of strains isolated from staphylococcalmastitis produce entero<strong>to</strong>xin A, which causes many human outbreaks.Several studies were successful in isolating the S. aureus phage type 80/81 fromskin lesions <strong>and</strong> cow’s milk, which is related <strong>to</strong> epidemic infections in man. One ofthe studies proved that phage type 80/81 produced interstitial mastitis in cows. Thesame phage type was found among animal caretakers, which indicates that thebacterium can be transmitted between man <strong>and</strong> animals <strong>and</strong> that the latter mayreinfect man.Infected fowl <strong>and</strong> dogs (see the section on occurrence in animals) may also giverise <strong>to</strong> <strong>and</strong> be a source of staphylococcal poisoning in man.One subject that deserves special attention is the appearance of antibiotic-resistantstrains in animals whose food contains antibiotics. There is concern regardingthe possible transmission of these strains <strong>to</strong> man. On several occasions, resistantstrains have been found both in animals (cows, swine, <strong>and</strong> fowl) <strong>and</strong> in their caretakers,with the same antibiotic resistance. Moreover, “human” strains (phage typed)have occasionally been isolated from the nostrils <strong>and</strong> lesions of other species ofdomestic animals.A variety of foods <strong>and</strong> dishes may be vehicles of the <strong>to</strong>xin. If environmental conditionsare favorable, S. aureus multiplies in the food <strong>and</strong> produces entero<strong>to</strong>xins.Once made, the <strong>to</strong>xin is not destroyed even if the food is subjected <strong>to</strong> boiling for theusual cooking time. Consequently, the <strong>to</strong>xin may be found in food while staphylococciare not.Poisoning is usually caused by primarily protein-based cooked foods that are contaminatedduring h<strong>and</strong>ling <strong>and</strong> left at room temperature. Red meat <strong>and</strong> fowl wereresponsible for 47.3% of the outbreaks in the US (ham was the most <strong>common</strong> sourcein that country) <strong>and</strong> 77.2% in Engl<strong>and</strong>. In Spain, primarily mayonnaise <strong>and</strong> foodscontaining mayonnaise were implicated; in Germany, four outbreaks were due <strong>to</strong>meat <strong>and</strong> three <strong>to</strong> eggs <strong>and</strong> milk products (Genigeorgis, 1989). During a Caribbeancruise, 215 of 715 passengers were poisoned by cream-filled pastries served at twodifferent meals on board. The remaining pastries were thrown out <strong>and</strong> could not bestudied, but entero<strong>to</strong>xigenic strains of S. aureus phage type 85/+ were isolated fromthe feces of 5 of 13 patients <strong>and</strong> from none of the controls. Isolates of the samephage were obtained from a perirectal sample <strong>and</strong> from a forearm lesion from twoof seven members of the ship’s crew who were in charge of pastry preparation(Waterman et al., 1987).An important causal fac<strong>to</strong>r in poisoning is keeping food at room temperature orinadequate refrigeration, practices which allow staphylococci <strong>to</strong> multiply. Lack of


STAPHYLOCOCCAL FOOD POISONING 255hygiene in food h<strong>and</strong>ling is another notable fac<strong>to</strong>r. Outbreaks of food poisoning mayoften be traced <strong>to</strong> a single dish.Role of Animals in the Epidemiology of the Disease: Most outbreaks are causedby human strains <strong>and</strong>, <strong>to</strong> a lesser degree, by strains from cattle or other animals.Animal products, such as meat, milk, cheese, cream, <strong>and</strong> ice cream, usually constitutea good substrate for staphylococcal multiplication. Pasteurization of milkdoes not guarantee safety if <strong>to</strong>xins were produced prior <strong>to</strong> heat treatment, as the <strong>to</strong>xinsare heat-resistant. Outbreaks have also been caused by reconstituted powderedmilk, even when the dried powder contained few or no staphylococci.Diagnosis: The short incubation period between ingestion of the food involved<strong>and</strong> the appearance of symp<strong>to</strong>ms is the most important clinical criterion. Labora<strong>to</strong>ryconfirmation, when possible, is based mainly on demonstration of the presence ofentero<strong>to</strong>xin in the food. Biological methods (inoculation of cats with cultures of thesuspect food, or of rhesus monkeys with the food or cultures) are expensive <strong>and</strong> notalways reliable. As substitutes, serological methods, such as immunodiffusion,immunofluorescence, hemagglutination inhibition, enzyme-linked immunosorbentassay (ELISA), <strong>and</strong> reverse passive latex agglutination are used (Windemann <strong>and</strong>Baumgartner, 1985; Shinagawa et al., 1990). These tests are useful in epidemiologicalresearch but not in daily practice (Benenson, 1990).The isolation of entero<strong>to</strong>xigenic staphylococcal strains from foods <strong>and</strong> typing byphage or immunofluorescence have epidemiological value. Quantitative examinationof staphylococci in processed or cooked foods serves as an indica<strong>to</strong>r of hygieneconditions in the processing plant <strong>and</strong> of personnel supervision.Control: Control measures include the following: (a) education of those who preparefood at home <strong>and</strong> other food h<strong>and</strong>lers, so that they will take proper personalhygiene measures; (b) prohibiting individuals with abscesses or other skin lesionsfrom h<strong>and</strong>ling food; (c) refrigeration at 4°C or lower of all foods in order <strong>to</strong> preventbacterial multiplication <strong>and</strong> the formation of <strong>to</strong>xins. Foods must be kept at roomtemperature for as little time as possible.The veterinary milk inspection service should supervise dairy installations, thecorrect operation of refrigeration units <strong>and</strong> their use immediately after milking, <strong>and</strong>refrigerated transport of the milk <strong>to</strong> pasteurization plants.The veterinary meat inspection service should be responsible for enforcinghygiene regulations before <strong>and</strong> after slaughter as well as during h<strong>and</strong>ling <strong>and</strong> processingof meat products. Control of hygiene conditions in meat retail establishmentsis also important.BibliographyBenenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bergdoll, M.S. The entero<strong>to</strong>xins. In: Cohen, J.O., ed. The Staphylococci. New York:Wiley; 1972.Bergdoll, M.S. Staphylococcal in<strong>to</strong>xications. In: Riemann, H., F.L. Bryan, eds. Food-BorneInfections <strong>and</strong> In<strong>to</strong>xications. 2nd ed. New York: Academic Press; 1979.


256 BACTERIOSESBergdoll, M.S., C.R. Borja, R.N. Robbins, K.F. Weiss. Identification of entero<strong>to</strong>xin E.Infect Immun 4:593–595, 1971.Bergdoll, M.S., B.A. Crass, R.F. Reiser, R.N. Robbins, J.P. Davis. A new staphylococcalentero<strong>to</strong>xin, entero<strong>to</strong>xin F, associated with <strong>to</strong>xic-shock-syndrome Staphylococcus aureus isolates.Lancet 1:1017–1021, 1981.Bergdoll, M.S., R. Reiser, J. Spitz. Staphylococcal entero<strong>to</strong>xin detection in food. FoodTechn 30:80–83, 1976.Bone, F.J., D. Bogie, S.C. Morgan-Jones. Staphylococcal food poisoning from sheep milkcheese. Epidemiol Infect 103:449–458, 1989.Casman, E.P., R.W. Bennett. Detection of staphylococcal entero<strong>to</strong>xin in food. ApplMicrobiol 13:181–189, 1965.Cohen, J.O., P. Oeding. Serological typing of staphylococci by means of fluorescent antibodies.J Bact 84:735–741, 1962.De Nooij, M.P., W.J. Van Leeuwen, S. Notermans. Entero<strong>to</strong>xin production by strains ofStaphylococcus aureus isolated from clinical <strong>and</strong> non-clinical specimens with special reference<strong>to</strong> entero<strong>to</strong>xin F <strong>and</strong> <strong>to</strong>xic shock syndrome. J Hyg 89:499–505, 1982.Evenson, M.L., M.W. Hinds, R.S. Bernstein, M.S. Bergdoll. Estimation of human dose ofstaphylococcal entero<strong>to</strong>xin A from a large outbreak of staphylococcal food poisoning involvingchocolate milk. Int J Food Microbiol 7:311–316, 1988.Fluharty, D.N. Staphylococcocis. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger,eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.Genigeorgis, C.A. Present state of knowledge on staphylococcal in<strong>to</strong>xication. Int J FoodMicrobiol 9:327–360, 1989.Gillespie, J.H., J.F. Timoney. Hagan <strong>and</strong> Bruner’s Infectious Diseases of DomesticAnimals. 7th ed. Ithaca: Coms<strong>to</strong>ck; 1981.Gross, E.M., Z. Weizman, E. Picard, et al. Milkborne gastroenteritis due <strong>to</strong> Staphylococcusaureus entero<strong>to</strong>xin B from a goat with mastitis. Am J Trop Med Hyg 39:103–104, 1988.Gutiérrez, L.M., I. Menes, M.L. García, et al. Characterization <strong>and</strong> entero<strong>to</strong>xigenicity ofstaphylococci isolated from mastitic ovine milk in Spain. J Food Protect 45:1282–1285, 1982.Hirooka, E.Y., E.E. Müller, J.C. Freitas, et al. Entero<strong>to</strong>xigenicity of Staphylococcus intermediusof canine origin. Int J Food Microbiol 7:185–191, 1988.Holmberg, S.D., P.A. Blake. Staphylococcal food poisoning in the United States. New facts<strong>and</strong> old misconceptions. JAMA 251:487–489, 1984.Kaji, Y., E. Ka<strong>to</strong>. Occurrence of entero<strong>to</strong>xigenic staphylococci in household <strong>and</strong> labora<strong>to</strong>rydogs. Jpn J Vet Res 28:86–94, 1980.Ka<strong>to</strong>, E., T. Kume. Entero<strong>to</strong>xigenicity of bovine staphylococci isolated from Californiamastitis test-positive milk in Japan. Jpn J Vet Res 28:75–85, 1980.Live, I. Staphylococci in animals: Differentiation <strong>and</strong> relationship <strong>to</strong> human staphylococcosis.In: Cohen, J.O., ed. The Staphylococci. New York: Wiley; 1972.Merchant, I.A., R.A. Packer. Veterinary Bacteriology <strong>and</strong> Virology. 7th ed. Ames: IowaState University Press; 1967.Mossel, D.A.A., F. Quevedo. Control microbiológico de los alimen<strong>to</strong>s. Lima: UniversidadNacional Mayor de San Marcos; 1967.Pulverer, G. Lebensmittelvergiftugen durch Staphylokokken. Bundesgesundhbl26:377–381, 1983.Raska, K., V. Matejosvska, L. Polak. To the origin of contamination of foodstuff by entero<strong>to</strong>xigenicstaphylococci. Proceedings of the World Congress of Foodborne Infections <strong>and</strong>In<strong>to</strong>xications. Berlin: Institute of Veterinary Medicine; 1980.Shinagawa, K., K. Watanabe, N. Matsusaka, et al. Enzyme-linked immunosorbent assay forthe detection of staphylococcal entero<strong>to</strong>xins in incriminated foods <strong>and</strong> clinical specimensfrom outbreaks of food poisoning. Jpn J Vet Sci 52:847–850, 1990.Thatcher, F.S., D.S. Clark. Análisis microbiológico de los alimen<strong>to</strong>s. Zaragoza:Acribia; 1972.


STREPTOCOCCOSIS 257Waterman, S.H., T.A. Demarcus, J.G. Wells, P.A. Blake. Staphylococcal food poisoning ona cruise ship. Epidemiol Infect 99:349–353, 1987.Windemann, H., E. Baumgartner. Bestimmung von Staphylokokken-Entero<strong>to</strong>xinen A, B, Cund D in Lebensmitteln mittels S<strong>and</strong>wich-ELISA mit markierten Antikörper. Zbl Bak Hyg IAbt Orig B 181:345–363, 1985.STREPTOCOCCOSIS(S. suis <strong>and</strong> other species of interest)ICD-10 A38 scarlet fever; G00.2 strep<strong>to</strong>coccal meningitis;J02.0 strep<strong>to</strong>coccal pharyngitisSynonyms: Strep<strong>to</strong>coccal infection, strep<strong>to</strong>coccal sore throat, scarlatina.Etiology: The genus Strep<strong>to</strong>coccus includes many species that display notabledifferences in their biological properties <strong>and</strong> their pathogenicity for man <strong>and</strong> animals.Strep<strong>to</strong>cocci are round, nonmotile, gram-positive bacteria that occur in pairsor long chains, particularly in fluid cultures. Strep<strong>to</strong>coccus does not form spores <strong>and</strong>certain species, such as S. suis, have capsules that can be seen when cultured inserum media.Lancefield’s serological classification is very useful for identifying these bacteria.This scheme currently distinguishes 20 serogroups <strong>and</strong> identifies them with the lettersA <strong>to</strong> V, excluding I <strong>and</strong> J. Many components of the serogroups have not beengiven specific names. Lancefield’s classification is based on a precipitation test withantisera for the different dominant polysaccharide antigens located on the bacteriumwall. Capsular species can in turn be divided in<strong>to</strong> serotypes. This is true of S. suis,which is currently subdivided in<strong>to</strong> 29 capsular serotypes (Higgins et al., 1992).Several serogroups produce additional antigens that serve <strong>to</strong> identify serotypes.Serotyping is useful in epidemiology.A single serogroup may include strains that are physiologically <strong>and</strong> biochemicallydifferent, thus classification cannot be based solely on serology (Timoney et al.,1988). Moreover, some strains cannot be typed serologically in a serogroup <strong>and</strong> canonly be identified on the basis of biochemical <strong>and</strong> physiological properties or by thecombination of these characteristics plus serology (Kunz <strong>and</strong> Moellering, 1981).A <strong>common</strong> technique for preliminary identification consists of dividing the strep<strong>to</strong>cocciin<strong>to</strong> three categories according <strong>to</strong> their hemolytic reactivity: alpha (incompletehemolysis <strong>and</strong> greenish discoloration on blood agar), beta (complete lysis oferythrocytes), <strong>and</strong> gamma (nonhemolytic). β-hemolytic strep<strong>to</strong>cocci are usually thecause of acute <strong>diseases</strong> <strong>and</strong> suppurative lesions, while α-hemolytic <strong>and</strong> Γ-strep<strong>to</strong>coccicause subacute disease, with some exceptions.S. suis serotype 2 is of particular interest in terms of <strong>zoonoses</strong> because transmissionfrom swine <strong>to</strong> man has been confirmed. This agent belongs <strong>to</strong> Lancefield’s


258 BACTERIOSESgroup D. There are other Strep<strong>to</strong>coccus species that are <strong>common</strong> <strong>to</strong> both man<strong>and</strong> animals, but which may or may not have specific reservoirs for differentanimal species.Geographic Distribution: Strep<strong>to</strong>cocci are distributed worldwide. S. suis isprobably prevalent in all areas where swine are bred.Occurrence in Man: Disease caused by S. suis in man is rare. Between 1968 <strong>and</strong>1984, it was isolated from 30 cases of meningitis in the Netherl<strong>and</strong>s; another 30cases caused by this agent occurred outside that country from 1968 <strong>to</strong> 1985 (Arends<strong>and</strong> Zanen, 1988).Infections caused by group A (S. pyogenes) are <strong>common</strong> in man, with an apparentlyhigher prevalence in temperate climates. For a long time, strep<strong>to</strong>cocci belonging<strong>to</strong> serogroup B (S. agalactiae) were considered mainly pathogenic for animals.They are now recognized as one of the major causes of septicemia, pneumonia, <strong>and</strong>meningitis in human newborns. In addition, strep<strong>to</strong>cocci belonging <strong>to</strong> serogroup D(S. bovis) are a frequent cause of endocarditis <strong>and</strong> bacteremia in man. There are sporadiccases of disease caused by strep<strong>to</strong>cocci belonging <strong>to</strong> groups C, G, F, H, <strong>and</strong>others. In man, there have been rare cases due <strong>to</strong> S. acidominimus, which is foundin milk <strong>and</strong> in the genital <strong>and</strong> intestinal tracts of cattle; <strong>to</strong> S. uberis, which causesmastitis in cows <strong>and</strong> is found in milk, the oropharynx, skin, <strong>and</strong> intestinal tract; <strong>to</strong>S. lactis <strong>and</strong> S. cremoris, which cause mastitis in cows <strong>and</strong> are found in cow milk;<strong>and</strong> <strong>to</strong> S. equi <strong>and</strong> its subspecies, S. zooepidemicus, which produce various <strong>diseases</strong>in animals. Finally, there is S. canis, groups G, L, <strong>and</strong> M (Gallis, 1990).Occurrence in Animals: Some <strong>diseases</strong> are very <strong>common</strong> <strong>and</strong> economicallyimportant. These include mastitis in cows caused by S. agalactiae (group B) <strong>and</strong>strangles caused by S. equi (group C) in horses <strong>and</strong> S. suis in swine.The Disease in Man: In the 60 cases recorded up <strong>to</strong> 1988, the clinically predominantform of infection caused by S. suis was meningitis. Most patients showedclassic symp<strong>to</strong>ms of meningitis: severe headache, high fever, confusion, <strong>and</strong> stiffneck. More than 50% experienced a loss of audi<strong>to</strong>ry acuity. Other complicationswere arthritis <strong>and</strong> endophthalmitis. Mortality was 7%. Most patients were employedin occupations involving the h<strong>and</strong>ling of swine or their products (swine breeders,slaughterhouse workers, butchers, swine transporters). Of the 30 patients in theNetherl<strong>and</strong>s, 28 cases were caused by S. suis type 2, 1 by type 4, <strong>and</strong> 1 by a strainthat could not be typed (Arends <strong>and</strong> Zanen, 1988). The same authors estimate thatin that country the risks for slaughterhouse workers <strong>and</strong> swine breeders would be 3per 100,000 inhabitants.S. pyogenes is the principal pathogen among hemolytic strep<strong>to</strong>cocci. This agentfrequently causes epidemics of septic sore throat <strong>and</strong> scarlet fever (strep<strong>to</strong>coccal<strong>to</strong>nsillitis <strong>and</strong> pharyngitis), various suppurative processes, septicemias, puerperalsepsis, erysipelas, ulcerative endocarditis, <strong>and</strong> other localized infections.Strep<strong>to</strong>coccal sore throat <strong>and</strong> scarlet fever are epidemiologically similar. The latteris differentiated clinically by the exanthema caused by strains producing an erythrogenic<strong>to</strong>xin. The disease is mild or inapparent in a high percentage of thoseinfected. Rheumatic fever is a sequela of strep<strong>to</strong>coccal sore throat or scarlet fever<strong>and</strong> may be caused by any strain of group A. Glomerulonephritis is another complication,produced only by certain nephri<strong>to</strong>genic strains of the same group.


STREPTOCOCCOSIS 259Group B strep<strong>to</strong>cocci are important causal agents of neonatal disease. Group Astrep<strong>to</strong>cocci <strong>and</strong> Staphylococcus aureus were replaced by Escherichia coli <strong>and</strong>serogroup B strep<strong>to</strong>cocci as the principal agents of neonatal infection. In infectionscaused by group B strep<strong>to</strong>cocci (S. agalactiae), two clinical syndromes are distinguished,depending on the age of the infant at the onset of disease. The acute orearly-onset syndrome appears between the first <strong>and</strong> fifth day of life <strong>and</strong> is characterizedby sepsis <strong>and</strong> respira<strong>to</strong>ry difficulty. The delayed-onset syndrome generallyappears after the tenth day <strong>and</strong> is characterized by meningitis, with or without sepsis.Affected children show lethargy, convulsions, <strong>and</strong> anorexia. Mortality is high inboth forms, but higher in the early-onset syndrome.In older children <strong>and</strong> adults, group B strep<strong>to</strong>cocci cause a variety of clinical syndromes:urinary tract infections, bacteremia, gangrene, postpartum infection, pneumonia,endocarditis, empyema, meningitis, <strong>and</strong> other pathological conditions(Patterson <strong>and</strong> el Ba<strong>to</strong>ol Hafeez, 1976).Disease caused by group C strep<strong>to</strong>cocci (S. equi) is sporadic <strong>and</strong> rare in man.However, in 1983, an epidemic outbreak occurred in New Mexico (USA), with 16cases caused by the consumption of white cheese made at home with unpasteurizedmilk. The agent was identified as S. zooepidemicus, one of the four species thatmake up group C. The disease in these patients consisted of fever, chills, <strong>and</strong> vagueconstitutional symp<strong>to</strong>ms, but five of them had a localized infection, which manifestedin such varied symp<strong>to</strong>ms as pneumonia, endocarditis, meningitis, pericarditis,<strong>and</strong> abdominal pains (CDC, 1983).In Engl<strong>and</strong> <strong>and</strong> Wales between 1983 <strong>and</strong> 1984, there were eight deaths during 32outbreaks associated with milk <strong>and</strong> milk products contaminated by S. zooepidemicus(Barrett, 1986). There were 11 cases in Hong Kong between 1982 <strong>and</strong> 1990 inpatients suffering from septicemia associated with a cardiovascular illness.Mortality was 22%. Five of the 11 patients had a predisposing disease. The sourceof infection was undercooked or raw pork (Yuen et al., 1990).In sporadic cases caused by strep<strong>to</strong>coccus group C, the most <strong>common</strong> clinicalmanifestation is exudative pharyngitis or <strong>to</strong>nsillitis. With some exceptions, group Cstrep<strong>to</strong>cocci isolated from these cases belong <strong>to</strong> S. equisimilis, which produces septicemiain suckling pigs. An outbreak of pharyngitis caused by group C strep<strong>to</strong>cocci,due <strong>to</strong> the consumption of raw milk, was followed by a high incidence of glomerulonephritis(Duca et al., 1969).Both enterococcal <strong>and</strong> nonenterococcal group D strep<strong>to</strong>cocci cause serious <strong>diseases</strong>in man. S. bovis causes bacteremia <strong>and</strong> endocarditis, <strong>and</strong> enterococci causeurinary tract infections, abdominal abscesses, <strong>and</strong> a significant percentage of casesof bacterial endocarditis. S. suis, already described, belongs <strong>to</strong> group D.Strep<strong>to</strong>cocci belonging <strong>to</strong> other serogroups, as well as those not serologicallygrouped, cause a wide variety of clinical manifestations, including dental caries <strong>and</strong>abscesses, meningitis, puerperal sepsis, wound infections, endocarditis, <strong>and</strong> otherpathological conditions (Kunz <strong>and</strong> Moellering, 1981).Nonhemolytic strep<strong>to</strong>cocci <strong>and</strong> “viridans” type (a-hemolytic) strep<strong>to</strong>cocci cancause subacute endocarditis.The preferred antimicrobial for treatment is penicillin (Benenson, 1990).The Disease in Animals: S. suis belongs <strong>to</strong> group D <strong>and</strong> can be β- or α-hemolytic(Timoney et al., 1988). This agent frequently causes septicemia, meningitis, pneu-


260 BACTERIOSESmonia, <strong>and</strong> arthritis. Less frequently, it causes endocarditis, polyserositis, encephalitis,<strong>and</strong> abscesses. Although the rate of infection in a herd can be high, it does notusually affect more than 5% (Clif<strong>to</strong>n-Hadley, 1984). Of 663 strains isolated from sickswine in Canada, 21% belonged <strong>to</strong> type 2 (the most frequently occurring type in allcountries), followed by types 1/2 (which has capsular antigens from 1 <strong>and</strong> 2) <strong>and</strong> 3,with 12% each. Types 20 <strong>and</strong> 26 were the only types not found (Higgins <strong>and</strong>Gottschalk, 1992). In Denmark, types 2 <strong>and</strong> 7 represented 75% of the isolates. Type7 was isolated more frequently than in other countries, usually in suckling pigsyounger than 3 weeks. Experimental inoculation with S. suis type 7 in suckling pigsunder 7 days old caused severe disease (Boetner et al., 1987). In Australia, type 1 hascaused septicemia, meningitis, <strong>and</strong> polyarthritis in suckling pigs (Cook et al., 1988).In weaned piglets from various regions of Australia, type 2 is predominant (Ossowiczet al., 1989), although types 3, 4, <strong>and</strong> 9 have also been isolated <strong>and</strong> there are indicationsthat they can produce the same disease picture. In another study in New SouthWales <strong>and</strong> Vic<strong>to</strong>ria (Australia), type 9 was predominant (Gogolewski et al., 1990).In cattle, sheep, <strong>and</strong> goats, strains of types 5 <strong>and</strong> 2 were isolated from purulentlesions in the lungs <strong>and</strong> from other extramammary sites (Hommez et al., 1988).Most isolates of S. suis type 2 are sensitive <strong>to</strong> penicillin.S. agalactiae (S. mastitidis), in Lancefield’s group B, is the principal agent ofchronic catarrhal mastitis in dairy cows. S. dysgalactiae (group C) <strong>and</strong> S. uberis(group E) cause sporadic cases of acute mastitis in bovines. S. pyogenes, a humanpathogen, can infect the cow’s udder, producing mastitis <strong>and</strong> leading <strong>to</strong> epidemicoutbreaks in man.Horse strangles, caused by S. equi (group C), is an acute disease of horses characterizedby inflammation of the pharyngeal <strong>and</strong> nasal mucosa, with a mucopurulentsecretion <strong>and</strong> abscesses of the regional lymph nodes.S. equisimilis (group C) infects different tissues in several animal species. GroupC strep<strong>to</strong>cocci that are adapted <strong>to</strong> animals <strong>and</strong> classified as S. zooepidemicus producecervicitis <strong>and</strong> metritis in mares <strong>and</strong> often cause abortions. They also cause septicemiain colts. They are pathogenic for bovines, swine, <strong>and</strong> other animals, in whichthey produce various septicemic processes.S. zooepidemicus (group C) is an opportunistic pathogen in many animal species.It is a commensal on the skin, the mucosa of the upper respira<strong>to</strong>ry tract, <strong>and</strong> in the<strong>to</strong>nsils of many animal species. In horses, it is the <strong>common</strong> agent of wound infections<strong>and</strong> is a secondary disease agent after a viral infection in the upper respira<strong>to</strong>rytract of colts <strong>and</strong> young animals. It is also the agent of other infections in horses(Timoney et al., 1988). In cows, S. zooepidemicus can cause acute mastitis when itenters a wound in the teat. A fatal case of septicemia was described in a chicken(Timoney et al., 1988).Strep<strong>to</strong>cocci belonging <strong>to</strong> other groups cause abscesses <strong>and</strong> different diseaseprocesses in several animal species. The many <strong>diseases</strong> caused by strep<strong>to</strong>cocci areclinically differentiated by the agent’s portal of entry <strong>and</strong> the tissue it affects.Source of Infection <strong>and</strong> Mode of Transmission: The reservoir of S. pyogenes isman. Transmission of this respira<strong>to</strong>ry disease agent (septic sore throat, scarlet fever)results from direct contact between an infected person, whether patient or carrier,<strong>and</strong> another susceptible person. The disease is most frequent among children from5 <strong>to</strong> 15 years old, but also occurs at other ages.


STREPTOCOCCOSIS 261In Germany, Denmark, the US, Great Britain, <strong>and</strong> Icel<strong>and</strong>, important epidemicshave had their origin in the consumption of raw milk or ice cream made with milkfrom cows with udders infected by S. pyogenes. These epidemics were due <strong>to</strong> infectionin the cows’ udders contracted from infected milkers. Between 1920 <strong>and</strong> 1944,103 such epidemics of septic sore throat <strong>and</strong> 105 of scarlet fever were recorded inthe US due <strong>to</strong> consumption of raw milk from cows with infected udders. In otherinstances, the milk was contaminated directly (without the udders’ being infected bypeople with septic sore throat or localized infections). In several epidemic outbreaks,the milk became contaminated after pasteurization.According <strong>to</strong> the <strong>WHO</strong> Expert Committee on Strep<strong>to</strong>coccal <strong>and</strong> StaphylococcalInfections (1968), contamination of milk products has caused small outbreaks ofstrep<strong>to</strong>coccal respira<strong>to</strong>ry disease, but these are increasingly less frequent.Pasteurization has been the most important fac<strong>to</strong>r in the reduction of strep<strong>to</strong>coccaloutbreaks resulting from milk. In Third World countries, much milk is still consumedraw, <strong>and</strong> even in developed countries, outbreaks are produced by productsmade with raw milk.Special attention has been given <strong>to</strong> neonatal sepsis caused by group B strep<strong>to</strong>cocci(S. agalactiae). Research has shown that S. agalactiae colonizes a high percentageof women (7% <strong>to</strong> 30% or more) in different locations, such as the intestinal tract, thecervicovaginal region, <strong>and</strong> the upper respira<strong>to</strong>ry tract. The agent is possibly transferredfrom the rectal region <strong>to</strong> the vaginal canal, since most of the bacteria are intestinal.Infants can become contaminated in utero or during childbirth. Only a smallpercentage of neonates (approximately 1%) become infected <strong>and</strong> fall ill; in most, theagent colonizes the skin <strong>and</strong> the mucosa without affecting their health. The principalvictims of the infection, especially in the case of the early-onset syndrome, arepremature infants, low birthweight babies, <strong>and</strong> those born after a difficult labor. Theprincipal reservoir of group B strep<strong>to</strong>cocci causing neonatal disease is clearly themother. The S. agalactiae serotypes isolated from mothers <strong>and</strong> sick newborns arealways the same. Although S. agalactiae is an agent of bovine mastitis <strong>and</strong> has alsobeen isolated from other animal species, there is no evidence that the infection istransmitted from animals <strong>to</strong> man. In general, human <strong>and</strong> animal strains differ insome biochemical, metabolic, <strong>and</strong> serologic properties. It has been experimentallyshown that human strains of S. agalactiae can produce mastitis in bovines (Patterson<strong>and</strong> el Ba<strong>to</strong>ol Hafeez, 1976). However, some studies have suggested that a percentageof human infections may have derived from a bovine source (Van den Heever<strong>and</strong> Erasmus, 1980; Berglez, 1981) or that there is reciprocal transmission betweenhumans <strong>and</strong> bovines. Nonetheless, research findings seem <strong>to</strong> indicate that if suchtransmission occurs, its importance is probably limited.The outbreak of disease caused by S. zooepidemicus (group C) in New Mexico(USA) (see the section on the disease in man) clearly indicates that raw milk <strong>and</strong>unpasteurized milk products can be the source of infection for man. The epidemiologicalinvestigation of this outbreak sampled milk from cows on the establishmentwhere the cheese was made as well as samples of the cheese itself. S. zooepidemicuswas isolated from many of the samples. In Europe, there have also been cases of S.zooepidemicus infection caused by ingestion of raw milk. A case of pneumonia causedby S. zooepidemicus in a woman who cared for a sick horse has been described (Roseet al., 1980). The cases of disease caused by S. zooepidemicus that occurred in HongKong were attributed <strong>to</strong> the consumption of cooked or raw pork (Yuen et al., 1990).


262 BACTERIOSESInfection caused by S. suis type 2 is a true zoonosis. It is a highly occupationaldisease among those who breed pigs or participate in slaughtering, processing, ormarketing them. Man contracts the infection primarily through skin lesions.The infection in swine is widespread in areas where these animals are bred. In anendemic herd, both sick <strong>and</strong> healthy pigs carry the agent in their nasal cavities <strong>and</strong><strong>to</strong>nsils. The percentage of carrier animals can reach 50% or more of the herd duringoutbreaks <strong>and</strong> fall <strong>to</strong> only 3% when there are no clinical cases. Carrier status can lastfor at least 45 days <strong>and</strong> may persist in animals treated with penicillin (Clif<strong>to</strong>n-Hadley <strong>and</strong> Alex<strong>and</strong>er, 1980). Among swine, the infection is transmitted through theair <strong>and</strong> possibly through the digestive route as well. Pigs can also be carriers of S.suis in the vaginal canal <strong>and</strong> piglets can become infected during delivery (Robertsonet al., 1991).Animals can also transmit groups G, L, <strong>and</strong> M <strong>to</strong> man, but the epidemiology ofthese cross-infections has not yet been elucidated.Role of Animals in the Epidemiology of the Disease: Swine are the reservoir<strong>and</strong> source of S. suis infection in man. Animals do not act as maintenance hosts forS. pyogenes, but can sometimes cause important epidemic outbreaks by contractingthe infection from man <strong>and</strong> retransmitting it by means of contaminated milk. Thereis no firm evidence that animals play any significant role in the transmission ofgroup B strep<strong>to</strong>cocci causing neonatal sepsis. Raw cow milk can be a source ofgroup C strep<strong>to</strong>coccal infection in humans.Diagnosis: If milk is suspected as the source of an epidemic outbreak in man, anattempt should be made <strong>to</strong> isolate the etiologic agent. Obviously, a correct identificationof the agent is required. From either a human or animal source, it is advisable<strong>to</strong> identify the serogroup of strep<strong>to</strong>cocci involved, <strong>and</strong> <strong>to</strong> establish the species wheneverpossible. However, few labora<strong>to</strong>ries have the human <strong>and</strong> material resourcesnecessary for this task.A method has been described for identifying pregnant women with heavy colonizationof the genital tract by group B strep<strong>to</strong>cocci (Jones et al., 1983). The goalof this procedure is <strong>to</strong> start treating the newborn with drugs immediately after birth<strong>to</strong> reduce morbidity <strong>and</strong> mortality due <strong>to</strong> neonatal sepsis caused by group Bstrep<strong>to</strong>cocci.Infection by S. suis should be suspected if the patient presents the clinical manifestationsdescribed <strong>and</strong> his or her occupation involves contact with swine or theirby-products. Culture, isolation, <strong>and</strong> typing can confirm the diagnosis.In swine, definitive diagnosis also depends on isolation <strong>and</strong> identification of theagent. In endemic herds, the symp<strong>to</strong>ma<strong>to</strong>logy may be sufficiently clear <strong>to</strong> make aclinical diagnosis during new outbreaks. In a study conducted in Quebec (Canada)with 1,716 weaned pigs belonging <strong>to</strong> 49 herds <strong>and</strong> 23 control herds, nasal <strong>and</strong> <strong>to</strong>nsilsamples were taken with swabs. The samples were cultured in a brain-heart infusionbroth, strengthened with a supplement selective for Strep<strong>to</strong>coccus <strong>and</strong> 5% anti-S. suis type 2 serum developed in goats. After the diameter of the precipitation zonewas measured in 539 isolates, serum plate agglutination was used <strong>to</strong> identify isolatesof S. suis serotype 2. This method successfully identified 93.1% of the cultures isolatedusing the diameter of the precipitation zone as the sole criterion. Specificitywas 94.5% <strong>and</strong> relative sensitivity was 88.7% (Moreau et al., 1989).


STREPTOCOCCOSIS 263Control: Those who work with swine or their by-products should pay attention<strong>to</strong> cuts or abrasions <strong>and</strong> treat them properly <strong>to</strong> prevent infection by S. suis type 2.As for preventing the disease in swine, there are doubts regarding the efficacy ofthe bacterins used against S. suis. However, many veterinarians <strong>and</strong> breeders maintainthat they prevent outbreaks of acute illness. Adding penicillin <strong>to</strong> feed whenpiglets are being weaned early can also control acute disease. The disadvantage isthat penicillin becomes inactive in feed (Fraser et al., 1991). Experimental testsshowed that tiamulin administered in water was effective in reducing the effects ofS. suis type 2 (Chengappa et al., 1990).Prevention of human infection transmitted through milk is achieved primarily bypasteurization. Infected persons should not participate in milking or h<strong>and</strong>ling milkor other foods.The prevention of neonatal sepsis has been attempted by active immunization ofpregnant women with capsular polysaccharides of group B strep<strong>to</strong>cocci, as well asby passive immunization with immunoglobulin preparations given intravenously.Both immunization methods are in the experimental stage. Promising results havebeen obtained with prophylactic intravenous administration of ampicillin <strong>to</strong> womenin labor. In this way, a significant level of the antibiotic is obtained in the amnioticfluid <strong>and</strong> in samples of the umbilical cord. Among the newborns of obstetric patientsreceiving this treatment, only 2.8% were colonized by group B strep<strong>to</strong>cocci <strong>and</strong>none became ill, while in the control group, 35.9% of the newborns were colonized<strong>and</strong> four developed the early-onset syndrome (Fischer et al., 1983).To reduce the prevalence of mastitis caused by S. agalactiae in dairy herds, cowstesting positive <strong>to</strong> the California Mastitis Test (CMT) are treated with penicillin byextramammary infusion. However, this procedure does not eradicate the infection,probably because of reinfection. Application of antiseptic creams <strong>to</strong> teat lesions canhelp <strong>to</strong> prevent mastitis caused by S. dysgalactiae <strong>and</strong> S. zooepidemicus. Bacterinshave been tried for preventing equine strangles caused by S. equi. Although theyconfer satisfac<strong>to</strong>ry immunity, they produce a local <strong>and</strong> systemic reaction (Timoneyet al., 1988).BibliographyArends, J.P., H.C. Zanen. Meningitis caused by Strep<strong>to</strong>coccus suis in humans. Rev InfectDis 10:131–137, 1988.Barrett, N.J. Communicable disease associated with milk <strong>and</strong> dairy products in Engl<strong>and</strong><strong>and</strong> Wales: 1983–1984. J Infect 12:265–272, 1986.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Berglez, I. Comparative studies of some biochemical properties of human <strong>and</strong> bovineStrep<strong>to</strong>coccus agalactiae strains. Zbl Bakt Hyg I Abst Orig 173:457–463, 1981.Boetner, A.G., M. Binder, V. Bille-Hansen. Strep<strong>to</strong>coccus suis infections in Danish pigs <strong>and</strong>experimental infection with Strep<strong>to</strong>coccus suis serotype 7. Acta Pathol Microbiol Sc<strong>and</strong> [B]95:233–239, 1987.Chengappa, M.M., L.W. Pace, J.A. Williams, et al. Efficacy of tiamulin against experimentallyinduced Strep<strong>to</strong>coccus suis type 2-infection in swine. J Am Vet Med Assoc197:1467–1470, 1990.


264 BACTERIOSESClif<strong>to</strong>n-Hadley, F.A. Studies of Strep<strong>to</strong>coccus suis type 2 infection in pigs. Vet Res Commun8:217–227, 1984.Clif<strong>to</strong>n-Hadley, F.A., T.J. Alex<strong>and</strong>er. The carrier site <strong>and</strong> carrier rate of Strep<strong>to</strong>coccus suistype II in pigs. Vet Rec 107:40–41, 1980.Cook, R.W., A.R. Jackson, A.D. Ross. Strep<strong>to</strong>coccus suis type 1 infection of suckling pigs.Aust Vet J 65:64–65, 1988.Davies, A.M. Diseases of man transmissible through animals. In: Van der Hoeden, J., ed.Zoonoses. Amsterdam: Elsevier; 1964.Duca, E., G. Teodorovici, C. Radu, A. Vita, P. Talasman-Niculescu, E. Bernescu, et al. Anew nephri<strong>to</strong>genic strep<strong>to</strong>coccus. J Hyg 67:691–698, 1969.Eickhoff, T.C. Group B strep<strong>to</strong>cocci in human infection. In: Wannamaker, L.W., J.M.Matsen, eds. Strep<strong>to</strong>cocci <strong>and</strong> Strep<strong>to</strong>coccal Diseases: Recognition, Underst<strong>and</strong>ing, <strong>and</strong>Management. New York: Academic Press; 1972.Fischer, G., R.E. Hor<strong>to</strong>n, R. Edelman. From the National Institute of Allergy <strong>and</strong> InfectiousDiseases. Summary of the National Institutes of Health workshop on group B strep<strong>to</strong>coccalinfection. J Infect Dis 148:163–166, 1983.Fluharty, D.M. Strep<strong>to</strong>coccosis. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger,eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.Fraser, C.M., J.A. Bergeron, A. Mays, S.E. Aiello, eds. The Merck Veterinary Manual. 7thed. Rahway: Merck; 1991.Gallis, H.A. Viridans <strong>and</strong> β-hemolytic (Non-Group A, B, <strong>and</strong> D) strep<strong>to</strong>cocci. In: M<strong>and</strong>ell,G.L., R.G. Douglas, Jr., J.E. Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rded. New York: Churchill Livings<strong>to</strong>ne, Inc.; 1990.Gogolewski, R.P., R.W. Cook, C.J. O’Connell. Strep<strong>to</strong>coccus suis serotypes associated withdisease in weaned pigs. Aust Vet J 67:202–204, 1990.Higgins, R., M. Gottschalk. Distribution of Strep<strong>to</strong>coccus suis capsular types in Canada in1991. Can Vet J 33:406, 1992.Hommez, J., J. Wullepit, P. Cassimon, et al. Strep<strong>to</strong>coccus suis <strong>and</strong> other strep<strong>to</strong>coccalspecies as a cause of extramammary infection in ruminants. Vet Rec 123:626–627, 1988.Jones, D.E., E.M. Friedl, K.S. Kanarek, J.K. Williams, D.V. Lim. Rapid identification ofpregnant women heavily colonized with group B strep<strong>to</strong>cocci. J Clin Microbiol 18:558–560, 1983.Kunz, L.J., R.C. Moellering. Strep<strong>to</strong>coccal infection. In: Balows, A., W.J. Hausler, Jr., eds.Diagnostic Procedures for Bacterial, Mycotic, <strong>and</strong> Parasitic Infections. 6th ed. Washing<strong>to</strong>n,D.C.: American Public Health Association; 1981.MacKnight, J.F., P.J. Ellis, K.A. Jensen, B. Franz. Group B strep<strong>to</strong>cocci in neonatal deaths.Appl Microbiol 17:926, 1969.Merchant, I.A., R.A. Packer. Veterinary Bacteriology <strong>and</strong> Virology. 7th ed. Ames: IowaState University Press; 1967.Moreau, A., R. Higgins, M. Bigras-Poulin, M. Nadeau. Rapid detection of Strep<strong>to</strong>coccussuis serotype 2 in weaned pigs. Am J Vet Res 50:1667–1671, 1989.Ossowicz, C.J., A.M. Poin<strong>to</strong>n, P.R. Davies. Strep<strong>to</strong>coccus suis isolated from pigs in SouthAustralia. Aust Vet J 66:377–378, 1989.Patterson, M.J., A. el Ba<strong>to</strong>ol Hafeez. Group B strep<strong>to</strong>cocci in human disease. Bacteriol Rev40:774–792, 1976.Robertson, I.D., D.K. Blackmore, D.J. Hampson, Z.F. Fu. A longitudinal study of naturalinfection of piglets with Strep<strong>to</strong>coccus suis types 1 <strong>and</strong> 2. Epidemiol Infect 107:119–126, 1991.Rose, H.D., J.R. Allen, G. Witte. Strep<strong>to</strong>coccus zooepidemicus (group C) pneumonia in ahuman. J Clin Microbiol 11:76–78, 1980.S<strong>to</strong>llerman, G.H. Strep<strong>to</strong>coccal disease. In: Beeson, P.B., W. McDermott, eds. Cecil-LoebTextbook of Medicine. 12th ed. Philadelphia: Saunders; 1967.


TETANUS 265Timoney, J.F., J.H. Gillespie, F.W. Scott, J.E. Barlough. Hagan <strong>and</strong> Bruner’s Microbiology<strong>and</strong> Infectious Diseases of Domestic Animals. 8th ed. Ithaca: Coms<strong>to</strong>ck; 1988.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Group C strep<strong>to</strong>coccal infections associated with eating homemadecheese—New Mexico. MMWR Morb Mortal Wkly Rep 32(39):510, 515–516, 1983.Van den Heever, L.W., M. Erasmus. Group B Strep<strong>to</strong>coccus—comparison of Strep<strong>to</strong>coccusagalactiae isolated from humans <strong>and</strong> cows in the Republic of South Africa. J S Afr Vet Assoc51:93–100, 1980.<strong>WHO</strong> Expert Committee on Strep<strong>to</strong>coccal <strong>and</strong> Staphylococcal Infections. Strep<strong>to</strong>coccal<strong>and</strong> Staphylococcal Infections. Report of a <strong>WHO</strong> Expert Committee. Geneva: <strong>WHO</strong>; 1968.(Technical Report Series 394).Yuen, K.Y., W.H. Se<strong>to</strong>, C.H. Choi, et al. Strep<strong>to</strong>coccus zooepidemicus (Lancefield group C)septicaemia in Hong Kong. J Infect 21:241–250, 1990.


TETANUSICD-10 A33 tetanus neona<strong>to</strong>rum; A34 obstetrical tetanus; A35 other tetanusSynonyms: Trismus, lockjaw.Etiology: Clostridium tetani; the pathology is produced by the neuro<strong>to</strong>xin of theinfectious agent, since the bacterium does not invade the animal body. C. tetani, likeall clostridia, is a gram-positive, anaerobic, motile bacillus, 2–2.5 microns long by0.3–0.5 microns in diameter. It forms terminal ovoid spores, giving it the appearanceof a tennis racket. While multiplying logarithmically, C. tetani amasses an intracellularneuro<strong>to</strong>xin called tetanospasmin, which is released when the cell lyses.Tetanospasmin is a very potent <strong>to</strong>xin. It is estimated that less than 2.5 ng/kg of bodyweightwould be fatal for man <strong>and</strong> that 0.3 ng/kg of bodyweight would be fatal fora guinea pig (Orenstein <strong>and</strong> Wassilak, 1991). Production of the neuro<strong>to</strong>xin is determinedby a plasmid gene (Finn et al., 1984).C. tetani spores are very resistant <strong>to</strong> environmental fac<strong>to</strong>rs <strong>and</strong> can survive in thesoil for many years.Geographic Distribution: Worldwide. The etiologic agent is a soil microorganismthat can also be found in the feces of animals <strong>and</strong> man. The spores of C. tetaniare found primarily in cultivated l<strong>and</strong> rich in organic matter, or in pastures. The diseaseoccurs more frequently in the tropics than in temperate or cold climates.Occurrence in Man: The incidence of the disease is low in industrialized countries;in developing countries, it still represents an important public health problem.In the decade 1951–1960, the mortality rate from tetanus was 0.16 per 100,000inhabitants in the US <strong>and</strong> Canada <strong>and</strong> 8.50 per 100,000 in Latin America, excluding


266 BACTERIOSESArgentina <strong>and</strong> Brazil. In 1987, it was estimated that 1,680,000 cases <strong>and</strong> 1,030,000deaths occurred worldwide. In 1973, 60% <strong>to</strong> 90% of cases occurred in newbornsduring the first month of life (Orenstein <strong>and</strong> Wassilak, 1991). Currently, the distributionof the disease by age group is completely different in the US. In the period1989–1990, there were 117 cases of tetanus in 34 states, with an annual incidenceof 0.02 per 100,000 inhabitants. In marked contrast <strong>to</strong> developing countries, 58% ofthe patients were 60 years of age or older <strong>and</strong> only one case occurred in a newborn.Case fatality bore a direct relationship <strong>to</strong> age: 17% in patients aged 40 <strong>to</strong> 49, <strong>and</strong>50% in those aged 80 or older (CDC, 1993).Inhabitants of rural areas are more exposed than those in urban areas. Case fatalityis high despite improved treatment.A study conducted in Paraguay demonstrated that tetanus is more frequent in menthan in women, <strong>and</strong> more <strong>common</strong> in newborns <strong>and</strong> children than in adults (VeraMartínez et al., 1976).In Argentina, the annual rates of incidence for the period 1965–1977 were 1.2 <strong>to</strong>1.7 per 100,000 inhabitants (except in 1967, when the rate was 3.1 per 100,000inhabitants). The disease was more frequent in subtropical or temperate provincesthan in the cold Patagonian provinces. Average hospital admissions for tetanus inBuenos Aires between 1968 <strong>and</strong> 1973 were higher during the hot months. Tetanusmortality in these municipal hospitals reached 35.8% <strong>and</strong> was eight times higher inchildren younger than 15 days than in other age groups (Mazzáfero et al., 1981).Table 4 shows the morbidity distribution by climate for tetanus in Argentina duringthe period 1967–1977. In 1990, 49 cases were reported; in 1991, there were 38 casesof all ages; <strong>and</strong> in 1992, there were 7 neonatal cases. Underreporting is evident,since the number of deaths exceeds the number of patients, as indicated by theauthorities in charge of the National Disease Surveillance System (Argentina,Ministerio de Salud y Acción Social, 1990, 1991, <strong>and</strong> 1992).Occurrence in Animals: The disease is infrequent in animals. There are enzooticareas, particularly in the tropics. Horses are the most susceptible species. Cases alsooccur in sheep <strong>and</strong> cattle.The Disease in Man: It is characterized by painful spasms of the masseter muscles(trismus) <strong>and</strong> neck muscles (rictus), but it frequently affects other muscles inthe body. Although the average incubation period is 14 days, it may vary from lessthan two days <strong>to</strong> several months. If the disease is not complicated by other infections,temperature may be normal or only slightly elevated. Reflexes are exaggerated,<strong>and</strong> rigidity of the abdominal muscles, urine retention, <strong>and</strong> constipation are<strong>common</strong>. The case fatality rate is high, but varies from one country <strong>to</strong> another. In theUS, fatality fell from 90% in 1947 <strong>to</strong> 60% in 1969. In 1989–1990, it was 17% inpatients aged 40 <strong>to</strong> 49 <strong>and</strong> 50% in those aged 80 or older. The disease is much moresevere when the incubation period is short <strong>and</strong> convulsions appear early. The longer,more frequent, <strong>and</strong> more intense the convulsions become, the worse the prognosis.The symp<strong>to</strong>ma<strong>to</strong>logy of neonatal tetanus is the same as that of the disease inadults; only the infection’s portal of entry differs. In newborns, the infection usuallyenters through the umbilical stump. At other ages, the route of entry is a wound.Puncture wounds produced by contaminated objects or trauma wounds are especiallydangerous. Surgical interventions <strong>and</strong> induced abortions performed withoutadequate asepsis have given rise <strong>to</strong> tetanus.


TETANUS 267Table 4. Distribution of tetanus morbidity according <strong>to</strong> political division<strong>and</strong> climate, Argentina, 1967–1977.Political Average number Population at middle Rate perdivision of notified of reporting period 100,000<strong>and</strong> climate cases per year (in thous<strong>and</strong>s) inhabitantsSubtropicalCatamarcaCorrientesChacoFormosaJujuyMisionesSaltaSantiago del EsteroTucumán168.83.419.238.514.26.715.320.415.732.64,2211755875722483234705335197943.91.93.36.75.62.13.33.83.04.1TemperateFederal DistrictBuenos AiresCórdobaEntre RíosLa PampaLa RiojaMendozaSan JuanSan LuisSanta Fe217.618.5111.920.916.53.40.64.53.11.536.219,4092,9749,2892,1778381771391,0254031872,2001.10.61.20.91.92.20.40.40.70.81.6ColdChubutRío NegroNeuquénSanta CruzTierra del Fuego3.70.61.31.60.2—76220228117094150.50.30.40.90.20.0Source: Bull Pan Am Health Organ 15:328, 1981.C. tetani is not an invasive bacteria. The spores enter through a wound that maybe an anaerobic medium, especially if there is tissular necrosis. Under such conditions,C. tetani enters a vegetative state, multiplies, <strong>and</strong> releases the neuro<strong>to</strong>xin as itlyses. The disease is due <strong>to</strong> tetanospasmin, a very potent neuro<strong>to</strong>xin (see the sectionon etiology). It enters the nervous system through the neuromuscular junctions ofalpha mo<strong>to</strong>r neurons. Tetanospasmin inhibits the release of various neurotransmitters,allowing the lower mo<strong>to</strong>r neurons <strong>to</strong> increase muscle <strong>to</strong>ne <strong>and</strong> produce convulsionssimultaneously in the agonist <strong>and</strong> antagonist muscles (Cate, 1990).The patient must be kept in an intensive care unit <strong>and</strong> treated with benzodiazepines<strong>to</strong> reduce anxiety, <strong>and</strong> <strong>to</strong> obtain a central anticonvulsive effect <strong>and</strong> muscularrelaxation. It is often necessary <strong>to</strong> continue with tracheal intubation or a tra-


268 BACTERIOSEScheos<strong>to</strong>my. Simultaneously with these measures, human antitetanus immunoglobulinmust be administered (intramuscular administration of 500 IU). Administrationof penicillin or other antibiotics is recommended <strong>to</strong> reduce the <strong>to</strong>xin load(Cate, 1990).The Disease in Animals: Horses are very susceptible <strong>to</strong> tetanus <strong>and</strong> usuallyacquire it from shoeing nails. They may also contract it from any other wound contaminatedwith C. tetani if anaerobic conditions favor its multiplication. Their symp<strong>to</strong>msare similar <strong>to</strong> those of human tetanus. Localized rigidity appears first, due <strong>to</strong><strong>to</strong>nic convulsions of the masseter muscles, the neck muscles, <strong>and</strong> the hind legs, followedby generalized rigidity. Reflexes are increased <strong>and</strong> the animals are easily startledby noise, which causes general convulsions.Postpartum cases are seen in cows, especially if the placenta is retained. Cattlehave a high rate of neutralizing antibodies against the neuro<strong>to</strong>xin (tetanospasmin) ofC. tetani,but the antibody level drops markedly after parturition, leaving the animalvery susceptible <strong>to</strong> the disease. In calves <strong>and</strong> lambs, tetanus often follows castration,especially when rubber b<strong>and</strong>s are used, since the necrotic tissue left by this operationfavors anaerobiosis.Dehorning, tail docking, <strong>and</strong> shearing may also give rise <strong>to</strong> the disease.Iatrogenic tetanus sometimes occurs after surgical operations <strong>and</strong> vaccinations.The incubation period lasts 2 <strong>to</strong> 14 days. The symp<strong>to</strong>ma<strong>to</strong>logy is similar <strong>to</strong> thatin man. Death occurs in 4 <strong>to</strong> 10 days.Treatment consists of tranquilizers, curariform agents, <strong>and</strong> 300,000 IU of tetanusanti<strong>to</strong>xin every 12 hours. Good results can be obtained in horses if they are treatedat the onset of the disease. The wound must also be cleaned <strong>and</strong> drained, <strong>and</strong> broadspectrum antibiotics administered (Fraser et al., 1991).Source of Infection <strong>and</strong> Mode of Transmission: The reservoir <strong>and</strong> source ofinfection is soil containing C. tetani. The etiologic agent is found in many soils, particularlycultivated soil rich in organic matter. Areas where the exposure risk is highare referred <strong>to</strong> as “telluric foci” of C. tetani.The agent is <strong>common</strong>ly found in horse feces. It has also been found in otherspecies, such as cattle, sheep, dogs, rats, <strong>and</strong> chickens; similarly, man may harbor C.tetani in the intestinal tract.Transmission is effected through wounds. Scabs or crusts promote multiplicationof the etiologic agent. Some cases are due <strong>to</strong> dog bites. Tetanospasmin is producedafter the spores have germinated, i.e., by the vegetative form of the bacteria.In Paraguay, of 2,337 cases studied from 1946 <strong>to</strong> 1972, the portal of entry was theumbilical stump in 31.7%, small wounds in 38.7%, wounds caused by removal ofthe chigoe flea Tunga penetrans in 7.7%, <strong>and</strong> the remainder followed induced abortions,surgical interventions, burns, <strong>and</strong> injections without proper asepsis (VeraMartínez et al., 1976).Role of Animals in the Epidemiology of the Disease: Tetanus is a disease<strong>common</strong> <strong>to</strong> man <strong>and</strong> animals, not a zoonosis. Some authors ascribe the role ofreservoir <strong>to</strong> animals (McComb, 1980; Benenson, 1990), but it is more likely thatthe disease agent derives from the soil, <strong>and</strong> that it is present in the digestive tract ofherbivores <strong>and</strong> omnivores only transi<strong>to</strong>rily <strong>and</strong> does not multiply there (Wilson <strong>and</strong>Miles, 1975; Smith, 1975). Nevertheless, domesticated animals can disseminate


TETANUS 269<strong>to</strong>xigenic strains of C. tetani by means of their feces, in cultivated as well as uncultivatedareas.Diagnosis: Prior existence of a wound <strong>and</strong> accompanying symp<strong>to</strong>ms are the basesfor diagnosis. Direct microscopic examination of wound material is useful. Giventhe urgency of diagnosis, the value of culturing C. tetani is doubtful. It is not alwayspossible <strong>to</strong> isolate the etiologic agent from a wound.Control: In man, given the soil origin of the infection, the only rational means ofcontrol is active immunization with <strong>to</strong>xoid. Children 2 <strong>to</strong> 3 months of age shouldreceive three doses of the <strong>to</strong>xoid in the triple DPT vaccine (diphtheria, pertussis,tetanus) at intervals of one month <strong>to</strong> six weeks. They should then receive a booster,preferably administered 18 months after the last dose. An initial series of three dosesinduces protective titers of anti<strong>to</strong>xin for 5 <strong>to</strong> 13 years in 90% or more of those vaccinated.Booster shots ensure higher titers of the anti<strong>to</strong>xin <strong>and</strong> probably conferimmunity throughout a woman’s childbearing years (Halsey <strong>and</strong> de Quadros, 1983).Periodic boosters of tetanus <strong>to</strong>xoid every 10 years are recommended, particularly forpopulation groups most at risk. The effectiveness of the <strong>to</strong>xoid was confirmed duringWorld War II. US soldiers who were vaccinated with three doses of tetanus <strong>to</strong>xoidexperienced one case of tetanus among 455,803 wounded, while in the unvaccinatedJapanese army, the incidence was 10 cases per 100,000 wounded soldiers.In developing countries, immunization is recommended for pregnant mothers <strong>to</strong>prevent tetanus mortality in newborns. The effectiveness of prenatal immunizationwith tetanus <strong>to</strong>xoid (ana<strong>to</strong>xin) has been demonstrated. Primary immunization consistsof administering two doses, one at the start of pregnancy <strong>and</strong> another onemonth later, but not beyond three weeks before birth. If a pregnant woman hasalready been immunized, she only needs a booster <strong>and</strong> probably has enough antibodies<strong>to</strong> protect the children she bears over the next five years (Stanfield <strong>and</strong>Galazka, 1984).Passive immunization with anti<strong>to</strong>xin should be reserved for persons with no previousactive immunization who must undergo surgical operations, as well as forwomen after abortion or birth <strong>and</strong> for their newborn children in high-risk areas. Theuse of human anti<strong>to</strong>xin serum is preferable, but if unavailable, horse or bovinehyperimmune serum can be used after the patient is tested for a possible allergicreaction <strong>to</strong> the serum.Wounds should be cleaned <strong>and</strong> debrided. Persons who have previously receivedbasic <strong>to</strong>xoid treatment should be given a booster if the wound is small <strong>and</strong> more than10 years have passed since the last dose. If the patient has a large, contaminatedwound, a booster <strong>to</strong>xoid should be given if he was not vaccinated in the last fiveyears. Persons who did not receive a full primary series of tetanus <strong>to</strong>xoid shouldreceive a dose of <strong>to</strong>xoid <strong>and</strong> may require an injection of human tetanusimmunoglobulin, if it is a major wound <strong>and</strong>/or is contaminated (Benenson, 1990).Control procedures in animals are similar. Horses in particular should be vaccinatedwith <strong>to</strong>xoid; two doses given one <strong>to</strong> two months apart are sufficient. If thehorse suffers from a potentially dangerous wound, another <strong>to</strong>xoid injection shouldbe given. If the animal has not received <strong>to</strong>xoid previously, 2,000 <strong>to</strong> 3,000 IU of anti<strong>to</strong>xinshould be given. At the same time, one dose of <strong>to</strong>xoid should be given <strong>and</strong>repeated one month later. The anti<strong>to</strong>xin confers passive immunity for approximatelytwo weeks. Colts are given <strong>to</strong>xoid at 2 months of age <strong>and</strong> mares are given <strong>to</strong>xoid in


270 BACTERIOSESthe last six weeks of pregnancy (Fraser et al., 1991). Operations such as dehorning,castration, <strong>and</strong> tail docking should be done in the most aseptic conditions possible<strong>and</strong> antiseptics should be applied <strong>to</strong> surgical wounds.Lambs in the first month of life can become passively immunized when the eweis vaccinated with two doses of aluminum phosphate-adsorbed <strong>to</strong>xoid. The firstinjection should be administered eight weeks <strong>and</strong> the second, three or four weeksbefore the birth (Cameron, 1983).BibliographyArgentina, Ministerio de Salud y Acción Social. Boletines Epidemiológicos Nacionales,1990, 1991, <strong>and</strong> 1992.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bytchenko, B. Geographical distribution of tetanus in the world, 1951–60. Bull WorldHealth Organ 34:71–104, 1966.Cameron, C.M., B.J. Van Biljon, W.J. Botha, P.C. Knoetze. Comparison of oil adjuvant <strong>and</strong>aluminium phosphate-adsorbed <strong>to</strong>xoid for the passive immunization of lambs against tetanus.Onderstepoort J Vet Res 50:229–231, 1983.Cate, T.R. Clostridium tetani (Tetanus). In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E. Bennett,eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: Churchill Livings<strong>to</strong>ne,Inc.; 1990.Cvjetanovic, B. Epidemiología del tétanos considerada desde un pun<strong>to</strong> de vista práctico desalud pública. Bol Oficina Sanit Panam 75:315–324, 1973.Finn, C.W., Jr., R.P. Silver, W.H. Habig, M.C. Hardegree, et al. The structural gene fortetanus neuro<strong>to</strong>xin is on a plasmid. Science 224:881–884, 1984.Fraser, C.M., J.A. Bergeron, A. Mays, S.E. Aiello, eds. The Merck Veterinary Manual. 7thed. Rahway: Merck; 1991.Halsey, N.A., C.A. de Quadros. Recent Advances in Immunization. Washing<strong>to</strong>n, D.C.: PanAmerican Health Organization; 1983. (Scientific Publication 451).Mazzáfero, V.E., M. Boyer, A. Moncayo-Medina. The distribution of tetanus in Argentina.Bull Pan Am Health Organ 15(4):327–332, 1981.McComb, J.A. Tetanus (Lockjaw). In: H. S<strong>to</strong>enner, W. Kaplan, M. Torten, eds. Vol 2,Section A: CRC H<strong>and</strong>book Series in Zoonoses. Boca Ra<strong>to</strong>n: CRC Press; 1980.Orenstein, W.A., S.G.F. Wassilak. Tetanus. In: Evans, A.S., P.S. Brachman, eds. BacterialInfection of Humans. 2nd ed. New York: Plenum Medical Book Co.; 1991.Rosen, H.M. Diseases caused by clostridia. In: Beeson, P.B., W. McDermott, J.B.Wyngaarden, eds. Cecil Textbook of Medicine. 15th ed. Philadelphia: Saunders; 1979.Rosen, M.N. Clostridial infections <strong>and</strong> in<strong>to</strong>xications. In: Hubbert, W.T., W.F. McCulloch,P.R. Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield:Thomas; 1975.Smith, J.W.G. Diphtheria <strong>and</strong> tetanus <strong>to</strong>xoids. Br Med Bull 25(2):177–182, 1969.Smith, L.D. Clostridial <strong>diseases</strong> of animals. Adv Vet Sci 3:465–524, 1957.Smith, L.D. The Pathogenic Anaerobic Bacteria. 2nd ed. Springfield: Thomas; 1975.Spaeth, R. Tetanus. In: Top, F.H., Sr., P.F. Wehrle, eds. Communicable <strong>and</strong> InfectiousDiseases. 7th ed. St. Louis: Mosby; 1972.Stanfield, J.P., A. Galazka. Neonatal tetanus in the world <strong>to</strong>day. Bull World Health Organ62:647–669, 1984.Tavares, W. Profilaxis do tetano. Fundamen<strong>to</strong>s e critica de sua realização. Rev Assoc MedBras 28:10–14, 1982.


TICK-BORNE RELAPSING FEVER 271United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Tetanus surveillance—United States, 1989–1990. MMWRMorb Mortal Wkly Rep 42:233, 1993.Vera Martínez, A., C.M. Ramírez Boettner, V.M. Salinas, R. Zárate. Tétanos: estudio clínicoy epidemiológico de 2.337 casos. Bol Oficina Sanit Panam 80:323–332, 1976.Wilson, G.S., A. Miles. Vol. 2. Topley <strong>and</strong> Wilson’s Principles of Bacteriology, Virology,<strong>and</strong> Immunity. 6th ed. Baltimore: Williams & Wilkins; 1975.World Health Organization (<strong>WHO</strong>). Guidelines for the prevention of tetanus. <strong>WHO</strong> Chron30:201–203, 1976.TICK-BORNE RELAPSING FEVERICD-10 A68.1Synonyms: Endemic relapsing fever, spiroche<strong>to</strong>sis, spirochetal fever, recurrenttyphus, borreliosis.Etiology: Spirochetes of the genus Borrelia (syn. Spirillum, Spirochaeta,Spironema). Given the close relationship of specificity between the tick species <strong>and</strong>the Borrelia strains it harbors, classification of the etiologic agent according <strong>to</strong> itsvec<strong>to</strong>r has been proposed. Thus, the agent transmitted by Ornithodoros hermsiiwould be named Borrelia hermsii, the one found in O. brasiliensis would be B.brasiliensis, etc. Other borreliae derive their species name from their geographicalregion of origin. These include B. hispanica,transmitted by O. erraticus; B. venezuelensi,transmitted by O. rudis; <strong>and</strong> B. caucasica, transmitted by O. verrucosus.However, not all researchers agree with this taxonomy. Some maintain that all thestrains adapted <strong>to</strong> different Ornithodoros species are merely variants of a singlespecies, Borrelia recurrentis, the agent of epidemic relapsing fever, transmitted bylice.Borreliae are helical bacteria 3–20 microns long by 0.2–0.5 microns in diameter.They are gram-negative, have flagella between the external <strong>and</strong> internal membranes,are actively motile, <strong>and</strong> change direction frequently. Some species (B. dut<strong>to</strong>ni, B.parkeri, B. turicata) grow in labora<strong>to</strong>ry culture media (Kelly, 1984).Geographic Distribution: Natural foci of Borrelia transmissible <strong>to</strong> man arefound worldwide, with the exception of Australia, New Zeal<strong>and</strong>, <strong>and</strong> Oceania.Occurrence in Man: The incidence is low. Man contracts the infection only uponentering the natural foci where infected Ornithodoros are found. In some regions ofAfrica, the vec<strong>to</strong>r O. moubata has become established in dwellings, where it lives indirt floors. In Latin America, O. rudis (O. venezuelensis) <strong>and</strong> O. turicata also havean affinity for dwellings.In 1969, the number of cases in South America was 278, with one death. In 1976,15 cases were reported in the US. Sporadic cases occur in the western US states, in


272 BACTERIOSESCanada (British Columbia), Mexico, Guatemala, Panama, Colombia, Venezuela,Ecuador, <strong>and</strong> Argentina.Although endemic relapsing fever is usually sporadic, at times group outbreaksoccur. In 1973, there was an outbreak with 62 cases (16 confirmed <strong>and</strong> 46 clinicallydiagnosed) among <strong>to</strong>urists at Gr<strong>and</strong> Canyon National Park in Arizona (USA) whowere lodged in rustic wooden cabins infested by rodents <strong>and</strong> their ticks. In 1976, anoutbreak occurred under similar circumstances in California, with 6 cases among 11<strong>to</strong>urists (Harwood <strong>and</strong> James, 1979).A telephone <strong>and</strong> mail survey was conducted of 10,000 people who visited theGr<strong>and</strong> Canyon. The results showed that there were 14 cases of relapsing feveramong the <strong>to</strong>urists, <strong>and</strong> that 7 of these had <strong>to</strong> be hospitalized. There was labora<strong>to</strong>ryconfirmation of 4 cases <strong>and</strong> clinical diagnosis of 10 cases. Rodent nests were foundbeneath the ceilings <strong>and</strong> underneath the floors of the cabins where the <strong>to</strong>urists werelodged. These nests may have sheltered the vec<strong>to</strong>rs of the infection, as frequentlyhappens with Ornithodoros (CDC, 1991).Occurrence in Animals: In natural foci, many wild animal species are infected,among them rodents, armadillos, opossums, weasels, tree squirrels, <strong>and</strong> bats.The Disease in Man: Epidemic relapsing fever (transmitted by lice) <strong>and</strong> endemicrelapsing fever (transmitted by ticks) have similar clinical pictures. The averageincubation period is 7 days after the tick bite, but may vary from 4 <strong>to</strong> 18 days. Thedisease is characterized by an initial pyrexia that lasts three <strong>to</strong> four days <strong>and</strong> begins<strong>and</strong> disappears suddenly. The fever, which may reach 41°C, is accompanied bychills, profuse sweating, vertigo, cephalalgia, myalgia, <strong>and</strong> vomiting. At times, erythemas,petechiae, epistaxis, <strong>and</strong> jaundice of varying degrees of severity may beobserved. After several days without fever, the attacks of fever recur several times,lasting longer than in the first episode. The primary characteristic of the disease isthe syndrome of periodic fevers. There are generally three <strong>to</strong> seven relapses of fever,with intervals of four <strong>to</strong> seven days (Barbour, 1990). Periodic recurrences are attributed<strong>to</strong> antigenic changes or mutations in the borreliae, against which the patientcannot develop immunity. Borreliae in the first attack are antigenically differentfrom those isolated in relapses <strong>and</strong> there is no protective immunity among theseserotypes. The variable antigens are proteins on the outer membrane <strong>and</strong> their variationis the result of a new DNA arrangement (Barbour, 1990).Treatment is based on tetracyclines. Complications consist of meningitis <strong>and</strong>some other neurological disorders, but these occur in a small percentage of patients.Endemic fever is fatal in 2% <strong>to</strong> 5% of cases.The Disease in Animals: Little is known about the natural course of the infection<strong>and</strong> its possible clinical manifestations in wild animals. As with many otherreservoirs of infectious agents in natural foci, the hosts <strong>and</strong> borreliae are probablywell adapted <strong>to</strong> each other, <strong>and</strong> the latter likely have little or no pathogenic effec<strong>to</strong>n their hosts.Borreliosis (spiroche<strong>to</strong>sis) of fowl is a serious disease in geese, ducks, <strong>and</strong> chickens.It is caused by B. anserina <strong>and</strong> transmitted by Argus persicus <strong>and</strong> A. miniatus.The bovine infection in South Africa produced by B. theileri <strong>and</strong> transmitted byMargaropus decoloratus <strong>and</strong> Rhipicephalus evertsi causes a benign disease. Theseborrelioses affect only animals <strong>and</strong> are not transmitted <strong>to</strong> man.


TICK-BORNE RELAPSING FEVER 273Figure 18. Tick-borne relapsing fever (Ornithodoros spp.).Mode of transmission.Wild animals(rodents, armadillos,skunks, weasels,squirrels, <strong>and</strong> bats)Tick biteTickOrnithodoros spp.BiteWild animals(rodents, armadillos,skunks, weasels,squirrels, <strong>and</strong> bats)By bites, uponentering natural fociManSource of Infection <strong>and</strong> Mode of Transmission (Figure 18): The borreliae thatcause endemic relapsing fever have as their reservoir wild animals <strong>and</strong> ticks of thegenus Ornithodoros; in addition, the latter are vec<strong>to</strong>rs of the infection. These ticksare xerophilic argasids that are long-lived <strong>and</strong> very resistant <strong>to</strong> dessication <strong>and</strong> longperiods of fasting in environments with low humidity <strong>and</strong> high temperatures.Borreliae survive in the ticks for a long time. Depending on the species ofOrnithodoros,transovarial transmission may vary from less than 1% <strong>to</strong> 100%. In theWestern Hemisphere, the most important vec<strong>to</strong>rs of Borrelia are O. hermsii, O. turicata,O. rudis, <strong>and</strong> possibly O. talaje. The continuous circulation of borreliae innature is ensured by the ticks’ characteristics <strong>and</strong> their feeding on infected wild animals.O. hermsii lives at altitudes of over 1,000 meters, feeds on the blood of squirrels,<strong>and</strong> can be found in rodent burrows <strong>and</strong> wooden huts. O. turicata attacks sheep<strong>and</strong> goats, as well as other animals, <strong>and</strong> infests hides, rodent <strong>and</strong> snake burrows, <strong>and</strong>pigsties.Transmission <strong>to</strong> humans is caused by a bite from an infected tick.Role of Animals in the Epidemiology of the Disease: Several species of wildanimals constitute the reservoir of the etiologic agent. The relative importance ofticks <strong>and</strong> wild animals as reservoirs is the subject of debate, but both undoubtedlyplay important roles in maintaining the infection in nature. An exception is infectionby B. dut<strong>to</strong>ni in Africa, which has not been found in animals <strong>and</strong> is transmitteddirectly <strong>to</strong> man by the tick O. moubata.Diagnosis: Diagnosis is based on demonstrating the presence of the etiologicagent in the patient’s blood during the febrile phase by dark-field microscopy usingfresh smears or films stained by Giemsa or Wright techniques, or by inoculation inmice. The number of borreliae diminishes or disappears at the end of a fever attack;


274 BACTERIOSESthus, intraperi<strong>to</strong>neal inoculation of young mice <strong>and</strong> examination of their blood 24 <strong>to</strong>72 hours after inoculation is advisable.Control: Control measures are difficult <strong>to</strong> apply <strong>and</strong> are impractical, since casesin the Western Hemisphere are rare <strong>and</strong> usually widely dispersed. The principal recommendationis <strong>to</strong> avoid being bitten by ticks living in caves, burrows of rodents <strong>and</strong>other animals, or primitive huts.Human dwellings should be built <strong>to</strong> keep out the hosts (rodents or others) ofOrnithodoros. In addition, the s<strong>to</strong>rage of wood inside or near buildings should beavoided. People entering natural foci should examine themselves for ticks periodically,in addition <strong>to</strong> using protective footwear <strong>and</strong> clothing. Repellents provide partialprotection; dimethyl phthalate is the most highly recommended.BibliographyBarbour, A.G. Antigenic variation of a relapsing fever Borrelia species. Ann Rev Microbiol44:155–171, 1990.Bruner, D.W., J.H. Gillespie. Hagan’s Infectious Diseases of Domestic Animals. 6th ed.Ithaca: Coms<strong>to</strong>ck; 1973.Coates, J.B., B.C. Hoff, P.M. Hoff, eds. Preventive Medicine in World War II. Vol. VII:Communicable Diseases: Arthropod-borne Diseases other than Malaria. Washing<strong>to</strong>n, D.C.:Department of the Army; 1964.Felsenfeld, O. Borreliae, human relapsing fever, <strong>and</strong> parasite-vec<strong>to</strong>r-host relationships.Bact Rev 29:46–74, 1965.Francis, B.J., R.S. Thompson. Relapsing fever. In: Hoeprich, P.D., ed. Infectious Diseases.Hagers<strong>to</strong>wn: Harper & Row; 1972.Geigy, R. Relapsing fevers. In: Weinmann, D., M. Ristic, eds. Vol 2: Infectious BloodDiseases of Man <strong>and</strong> Animals. New York: Academic Press; 1968.Harwood, K.F., M.T. James. En<strong>to</strong>mology in Human <strong>and</strong> Animal Health. 7th ed. New York:Macmillan; 1979.Jellison, W.J. The endemic relapsing fevers. In: Hubbert, W.T., W.F. McCulloch, P.R.Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield:Thomas; 1975.Kelly, R.T. Genus IV Borrelia. In: Krieg, N.R., J.G. Holt, eds. Vol 1: Bergey’s Manual ofSystematic Bacteriology. Baltimore: Williams & Wilkins; 1984.Pan American Health Organization. Reported Cases of Notifiable Diseases in the Americas,1969. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1972. (Scientific Publication 247).United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Outbreak of relapsing fever—Gr<strong>and</strong> Canyon National Park,Arizona, 1990. MMWR Morb Mortal Wkly Rep 40(18):296–297, 303, 1991.


TULAREMIA 275TULAREMIAICD-10 A21.0 ulcerogl<strong>and</strong>ular tularaemia; A21.1 oculogl<strong>and</strong>ular tularaemia;A21.2 pulmonary tularaemia; A21.7 generalized tularaemia;A21.8 other forms of tularaemiaSynonyms: Francis’ disease, deer-fly fever, rabbit fever, Ohara’s disease.Etiology: Francisella tularensis, a highly pleomorphic, gram-positive, nonmotilebacillus; it has a fine capsule <strong>and</strong> can survive for several months in water, mud, <strong>and</strong>decomposing cadavers.Two biovars are recognized: F. tularensis biovar tularensis (Jellison type A) <strong>and</strong> F.tularensis biovar palaearctica (Jellison type B). Names have also been suggested forsome local biovars, such as mediaasiatica (Olsufjev <strong>and</strong> Meshcheryakova, 1982) <strong>and</strong>japonica. Classification in<strong>to</strong> biovars is not based on antigenic differences, but on theagent’s biochemical, virulence, <strong>and</strong> ecologic characteristics, <strong>and</strong> its nosography.Geographic Distribution: Natural foci of infection are found in the NorthernHemisphere. In the Americas, the disease has been confirmed in Canada, the US, <strong>and</strong>Mexico. It is found in most European countries, Tunisia, Turkey, Israel, Iran, China,<strong>and</strong> Japan. In the former Soviet Union, there are extensive areas with natural foci.F. tularensis biovar tularensis predominates in North America <strong>and</strong> causes 70% <strong>to</strong>90% of human cases in that part of the world. The principal sources of infection bythis biovar are lagomorphs (mainly those of the genus Sylvilagus) <strong>and</strong> ticks. Biovarpalaearctica (syn. holarctica) causes 10% <strong>to</strong> 30% of human cases; its principalhosts are rodents. Biovar tularensis is more virulent than biovar palaearctica (Bell<strong>and</strong> Reilly, 1981).Biovar palaearctica is found in western <strong>and</strong> northern Europe, Siberia, theFar East, in some parts of central Europe, <strong>and</strong> less frequently, in North America.Biovar palaearctica is distributed in natural foci among Rodentia spp. <strong>and</strong>Lagomorpha spp. In the Asian part of the former Soviet Union, where there are naturalfoci among Lepus <strong>and</strong> Gerbilinae, the name F. tularensis var. mediaasiaticahas been suggested for the etiologic agent. This biovar, like palaearctica, is moderatelyvirulent. Genetic studies have shown that the mediaasiatica <strong>and</strong> japonicavarieties hybridize with F. tularensis var. tularensis, indicating the possibility ofgenetically related strains outside of North America. The strains of Central Asiadiffer from the two main biovars in their glucose fermentation properties(S<strong>and</strong>ström et al., 1992).Occurrence in Man: It is not an internationally reportable disease <strong>and</strong> its globalincidence is hard <strong>to</strong> establish. The countries with the best data are the US <strong>and</strong> theformer Soviet Union. In both, the number of human cases has apparently declinedsharply. In the former Soviet Union, where in the 1940s some 100,000 cases werereported annually, the incidence has diminished <strong>to</strong> a few hundred cases per year. Inthe US, the average number of annual cases fell from 1,184 in the 1940s <strong>to</strong> some274 cases between 1960 <strong>and</strong> 1969 <strong>and</strong> has continued <strong>to</strong> fall.In the period 1977–1986, the average number of cases per year was 225. Manymild cases are not reported (Rohrbach, 1988). Current incidence is approximately


276 BACTERIOSES0.6 <strong>to</strong> 1.3 per million inhabitants (Boyce, 1990). The reduced incidence is attributed<strong>to</strong>, among other fac<strong>to</strong>rs, limited dem<strong>and</strong> for beaver (Cas<strong>to</strong>r canadensis) <strong>and</strong> muskrat(Ondatra zibethicus) skins <strong>and</strong> the resulting decline in hunting for these animals. Inthe US, 50% or more of the cases appear in a few states, such as Arkansas, Missouri,Oklahoma, Tennessee, <strong>and</strong> Texas. Although sporadic cases have occurred in allstates except Hawaii, their numbers have fallen.In areas where transmission is effected primarily by arthropods, incidence peaksin spring <strong>and</strong> summer. In contrast, in areas where cases of infection transmitted bywild rabbits predominate, the peaks occur in winter (Boyce, 1990), during the huntingseason.Epidemiological data on 1,026 human cases in the midwestern US states indicatedthat 63% involved an attached tick <strong>and</strong> 23% exposure <strong>to</strong> wild rabbits or other animals,such as squirrels, cats, <strong>and</strong> raccoons (Taylor et al., 1991). In Canada, therewere 31 cases between 1975 <strong>and</strong> 1979 (Akerman <strong>and</strong> Embil, 1982).Occurrence in Animals: The disease affects a large number of vertebrates (morethan 100 species of wild <strong>and</strong> domestic animals) <strong>and</strong> invertebrates (more than 100species). Natural infection has been found in ticks, mosqui<strong>to</strong>es, horseflies, fleas, <strong>and</strong>lice that parasitize lagomorphs <strong>and</strong> rodents.Epizootic outbreaks have been described in sheep, commercially bred furbearers(mink, beaver, <strong>and</strong> fox), <strong>and</strong> wild rodents <strong>and</strong> lagomorphs.In Sweden, epizootics occur in hares (Lepus timidus), the principal source ofhuman infection caused by F. tularensis biovar palaearctica. Between 1973 <strong>and</strong>1985, 1,500 samples were submitted <strong>to</strong> the National Veterinary Institute in Uppsala,divided nearly equally in<strong>to</strong> Lepus europeus <strong>and</strong> L. timidus. Tularemia was diagnosedby immunofluorescence in 109 samples of L. timidus,but in none of the L. europeussamples. The rate of animals infected varied by year; the highest rates occurred inautumn (Mörner et al., 1988).Few serological surveys have been conducted in domestic animals (Rohrbach,1988). In endemic areas of Georgia <strong>and</strong> Florida, titers of ≥ 1/80 were found in 2(6.2%) of 32 stray cats. As a result of an outbreak of 12 human cases of tularemiaon the Crow Indian reservation in southern Montana (USA), 90 dogs were testedserologically. Of these, 56 had agglutination titers of ≥ 1/40, whereas in a nearby<strong>to</strong>wn, only 6 of 34 yielded similar titers (Schmid et al., 1983).A study conducted in western Georgia <strong>and</strong> northwestern Florida (USA) used theserum agglutination test on 2,004 mammals of 13 species; 344 animals of 10 specieswere positive with titers of ≥ 1/80 (McKeever et al., 1958).The Disease in Man: It is seen most <strong>common</strong>ly as sporadic cases, but epidemicoutbreaks have occurred in the US <strong>and</strong> the former Soviet Union.The incubation period usually lasts from three <strong>to</strong> five days, but may range fromone <strong>to</strong> ten days. Several clinical forms of the disease are known; they are determinedprincipally by the agent’s route of entry. In all its forms, the disease is of suddenonset, with rising <strong>and</strong> falling fever, chills, asthenia, joint <strong>and</strong> muscle pain, cephalalgia,<strong>and</strong> vomiting. The most <strong>common</strong> clinical form is ulcerogl<strong>and</strong>ular, which represents85% of all cases in the Western Hemisphere. A local lesion is seen at the siteof entry (an arthropod bite, or a scratch or cut inflicted by contaminated nails orknife), which progresses <strong>to</strong> a necrotic ulceration accompanied by swelling of thenearby lymph node. The node frequently suppurates, ulcerates, <strong>and</strong> becomes scle-


TULAREMIA 277rotic. In untreated cases, the disease course lasts three <strong>to</strong> five weeks <strong>and</strong> convalescencetakes several weeks or months, with intermittent bouts of fever. A variety ofthis form is the gl<strong>and</strong>ular, in which there is no primary lesion; this is the most prevalenttype in Japan. The oculogl<strong>and</strong>ular form develops when contaminated materialcomes in<strong>to</strong> contact with the conjunctiva. The primary lesion localizes on the lowereyelid <strong>and</strong> consists of an ulcerated papule; at the same time, the regional lymphnodes swell. The primary pulmonary form, caused by aerosols, affects rural <strong>and</strong> labora<strong>to</strong>ryworkers, <strong>and</strong> produces pneumonia in one or both lungs. The typhoidal formis rare; it is caused by ingestion of contaminated foods (usually infected wild rabbitmeat) or water. It is a systemic disease that has very varied symp<strong>to</strong>ms <strong>and</strong> is difficult<strong>to</strong> diagnose. It is sometimes expressed as gastroenteritis, fever, <strong>and</strong> <strong>to</strong>xemia.Pneumonia is frequent in typhoidal tularemia. If not treated early, the course of thisclinical form may be short <strong>and</strong> fatal. Mortality in the pulmonary forms is high. Prior<strong>to</strong> the existence of antibiotics, mortality for all cases of tularemia in the US wasclose <strong>to</strong> 7%. Mortality outside of the Americas has rarely exceeded 1%. This differenceis attributed <strong>to</strong> the greater virulence of the tick-transmitted strains of F. tularensis(biovar tularensis) in the US. In the former Soviet Union, untreated cutaneousinfections (ulcerogl<strong>and</strong>ular form) are fatal for less than 0.5% of patients (biovarpalaearctica).The results of serologic <strong>and</strong> skin sensitivity tests carried out among exposedgroups show that inapparent infections are <strong>common</strong>.Strep<strong>to</strong>mycin is the preferred antibiotic for all forms of tularemia. Recommendedtreatment is 15 <strong>to</strong> 20 mg/kg/day of strep<strong>to</strong>mycin via intramuscular administration,divided in<strong>to</strong> various doses over 7 <strong>to</strong> 14 days (Boyce, 1990). Labora<strong>to</strong>ry tests withSc<strong>and</strong>inavian strains of F. tularensis biovar palaearctica obtained the lowest minimalinhibi<strong>to</strong>ry concentration with quinolones, as compared <strong>to</strong> other antibiotics. Thisresult should be taken in<strong>to</strong> account in clinical assays. In the Sc<strong>and</strong>inavian countries,where most tularemia patients are ambula<strong>to</strong>ry, quinolones have the additionaladvantage that they can be administered orally (Scheel et al., 1993).The Disease in Animals: In has been demonstrated experimentally that susceptibility<strong>to</strong> F. tularensis varies in different species of wild animals. Three groups havebeen established based on the infecting dose <strong>and</strong> the lethal dose. Group 1, the mostsusceptible, contains most species of rodents <strong>and</strong> lagomorphs, which generally suffera fatal septicemic disease. Group 2 is composed of other species of rodents <strong>and</strong>birds, which though highly susceptible <strong>to</strong> the infection, rarely die from it. Group 3consists of carnivores, which require high doses <strong>to</strong> become infected, rarely developbacteremia, <strong>and</strong> only occasionally manifest overt disease.Group 1 animals are an important source of infection for arthropods, other animals,man, <strong>and</strong> the environment. The clinical picture of the natural disease in theseanimals is not well known, since they are usually found dead or dying.Experimentally inoculated hares show weakness, fever, ulcers, abscesses at the inoculationsite, <strong>and</strong> swelling of the regional lymph nodes. Death ensues in 8 <strong>to</strong> 14 days.The lesions resemble those of plague <strong>and</strong> pseudotuberculosis, with caseous lymphnodes <strong>and</strong> grayish white foci in the spleen.High-mortality outbreaks have occurred in sheep in enzootic areas in Canada, theUS, <strong>and</strong> the former Soviet Union. In addition <strong>to</strong> causing economic losses, tularemiain sheep is a source of infection for man. In the US, the infection is transmitted by


278 BACTERIOSESthe tick Dermacen<strong>to</strong>r <strong>and</strong>ersoni, which during outbreaks is found in great numbersat the base of the sheep’s ears <strong>and</strong> on the neck. Sick animals separate themselvesfrom the flock <strong>and</strong> manifest fever, rigid gait, diarrhea, frequent urination, <strong>and</strong> respira<strong>to</strong>rydifficulty. Most deaths occur among young animals. Pregnant ewes mayabort. Reactions <strong>to</strong> serologic tests indicate that many animals have an inapparentinfection. Sheep can be classified in group 2 based on their susceptibility <strong>to</strong> theinfection. Au<strong>to</strong>psy reveals infarcts of the regional lymph nodes, mainly those of thehead <strong>and</strong> neck, as well as pneumonic foci. In this species, tularemia is a seasonaldisease, coinciding with tick infestations.The disease has been confirmed on occasion in horses, with symp<strong>to</strong>ms thatinclude lack of coordination, fever, <strong>and</strong> depression. The animals were parasitized bya large number of ticks. Infected young swine can manifest fever, dyspnea, <strong>and</strong>depression. Cattle seem <strong>to</strong> be resistant (Rohrbach, 1988).Cats can become infected <strong>and</strong> fall ill when hunting rodents in endemic areas or byconsuming dead lagomorphs. Cats can, in turn, transmit the infection <strong>to</strong> man. In acase that occurred in Georgia (USA), a young man who contracted ulcerogl<strong>and</strong>ulartularemia had three Siamese cats that had fallen ill two weeks earlier. The cats hadfever, anorexia, <strong>and</strong> apathy; the veterinarian prescribed strep<strong>to</strong>mycin <strong>and</strong> penicillin.The animals were cared for by their owner, who developed a necrotic ulcerous lesionthat started with a wound on his finger, although he did not recall having beenscratched or bitten. The three cats died despite treatment. On au<strong>to</strong>psy, necrotic fociwere found in the liver <strong>and</strong> spleen that contained coccobacilli positive for F. tularensiswith immunofluorescence.Another case was described in New Mexico (USA). The patient found his catunder the bed eating a dead wild rabbit. He tried <strong>to</strong> remove the cat <strong>and</strong> was bitten;four days later he fell ill with tularemia. The cat fell ill one day earlier, with apathy,anorexia, <strong>and</strong> fever, but no other symp<strong>to</strong>ms. The veterinarian did not prescribe anytreatment <strong>and</strong> the animal was found <strong>to</strong> be healthy when examined a week later.Serum agglutination yielded a titer of 1/160. The owner also recovered, after beingtreated with strep<strong>to</strong>mycin (CDC, 1982). In Oklahoma (USA), a state consideredendemic, a case of acute tularemia in three cats was diagnosed clinically <strong>and</strong> thenconfirmed by culture <strong>and</strong> immunofluorescence. The three animals showed signs ofdepression, lethargy, ulcerated <strong>to</strong>ngue <strong>and</strong> palate, moderate lymphadenomegaly,hepa<strong>to</strong>splenomegaly, <strong>and</strong> panleukopenia, with a severe <strong>to</strong>xic change in the neutrophils.Upon necropsy, multiple necrotic foci were found in the lymph nodes, liver,<strong>and</strong> spleen, as well as severe enterocolitis. The diagnosis was confirmed byimmunofluorescence <strong>and</strong> culture (Baldwin et al., 1991). Although tularemia is rarein cats, it should be kept in mind in enzootic areas. Since 1928, only 51 human caseshave been described that involve exposure <strong>to</strong> infected cats (Capellan <strong>and</strong>Fong, 1993).Source of Infection <strong>and</strong> Mode of Transmission (Figure 19): In natural foci, theinfection circulates among wild vertebrates, independently of man <strong>and</strong> domestic animals.Ticks are biological vec<strong>to</strong>rs of F. tularensis; not only do they transmit the etiologicagent from donor animals <strong>to</strong> other animals, they also constitute an importantinterepizootic reservoir. They are also responsible for transtadial <strong>and</strong> transovarialtransmission of the bacteria. Each enzootic region has one or more species of vertebrateanimals <strong>and</strong> of ticks that play the primary roles of transmitting <strong>and</strong> maintain-


TULAREMIA 279Figure 19. Tularemia. Mode of transmission in the Americas.Infected wildanimals: septicemicwild lagomorphs <strong>and</strong>rodents (Sylvilagus spp.,Cas<strong>to</strong>r canadensis,muskrats,Ondatra zibethicus)Tick biteTicksBiteSusceptible wildanimals: septicemicwild lagomorphs <strong>and</strong>rodents (Sylvilagus spp.,Cas<strong>to</strong>r canadensis,muskrats,Ondatra zibethicus)H<strong>and</strong>ling game animals,ingestion of contaminatedwater <strong>and</strong> meat, aerosolsBiteBiteBiteManSheepManDogsing the infection in nature. It is a matter of debate whether very susceptible lagomorphs<strong>and</strong> rodents (group 1) are true reservoirs or only amplifiers <strong>and</strong> the mainsource of infection for man. Less susceptible animals (group 2), <strong>to</strong>gether with ticks,are thought <strong>to</strong> be important reservoirs.Domestic animals, such as sheep <strong>and</strong> cats, are accidental hosts, but they may alsoconstitute sources of infection for man.Humans contract the infection upon entering the natural foci of tularemia. Thesources of infection <strong>and</strong> modes of transmission of the causal agent are many. InNorth America, the animals that most frequently serve as the source of infection forman are wild rabbits (Sylvilagus spp.), hares (Lepus californicus), beavers (Cas<strong>to</strong>rcanadensis), muskrats (Ondatra zibethicus), meadow voles (Microtus spp.), <strong>and</strong>sheep. The biovar tularensis is generally transmitted by wild rabbits or by their ticks(Dermacen<strong>to</strong>r variabilis, D. <strong>and</strong>ersoni, Amblyomma americanum). The biovarpalaearctica is more <strong>common</strong> among rodents, particularly aquatic rodents, but alsoin some species of lagomorphs, such as Lepus europaeus <strong>and</strong> L. variabilis.Tularemia in Sweden is transmitted from L. variabilis by means of mosqui<strong>to</strong>es. TheEuropean hare plays no role in Sweden, but it does in other European countries.Rodents such as beavers <strong>and</strong> muskrats are important in aquatic cycles. In differentecological areas, other ticks (e.g., Ixodes spp., Haemophysalis spp.) <strong>and</strong> arthropodsare also involved. In many enzootic areas, the principal route of penetration isthrough the skin (by means of hema<strong>to</strong>phagous arthropods, scratches, or knife cuts).Another portal of entry is the conjunctiva, which can be contaminated by materialssplashed in<strong>to</strong> the eyes or, in the case of hunters or sheep shearers, by h<strong>and</strong>s soiledfrom h<strong>and</strong>ling sick animals. Infection via the oral route occurs as a result of ingestingwater contaminated by dead animals or the urine <strong>and</strong> feces of infected animals,or by eating undercooked meat of lagomorphs or other infected animals. In addition,


280 BACTERIOSESthe disease can be contracted through the respira<strong>to</strong>ry system by inhaling aerosolscontaminated in the labora<strong>to</strong>ry or dust from fodder, grain, or wool contaminatedwith rodent excreta.Some cases of human infection by cat scratches or bites have been described. It isassumed that these animals had recently hunted <strong>and</strong> captured sick rodents or hadeaten dead lagomorphs. Another case occurred in a person exposed <strong>to</strong> a cat with anulcer (CDC, 1982). The disease was also contracted by a Canadian zoo veterinarianwho was bitten on the finger when treating a sick primate (Sanguinus nigricollis). Inthis zoo, four primates in adjacent cages died from tularemia, possibly transmitted byfleas from squirrels that often came near the cages. F. tularensis was isolated fromone of the squirrels. The primate responsible for infecting the veterinarian had sialorrhea,ocular <strong>and</strong> nasal discharges, <strong>and</strong> ulcers on the <strong>to</strong>ngue (Nayar et al., 1979).The highest incidence of cases occurs in the summer, when ticks are most active.Hunters are an especially vulnerable group, <strong>and</strong> the number of human casesincreases in hunting season.Role of Animals in the Epidemiology of the Disease: Human-<strong>to</strong>-human transmissionhas not been confirmed. Tularemia is a zoonosis that is transmitted <strong>to</strong> man(an accidental host) through contact with wild or domestic animals (of the latter,usually sheep), by a contaminated environment, or by such vec<strong>to</strong>rs as ticks, horseflies,<strong>and</strong> mosqui<strong>to</strong>es.Diagnosis: In man, clinical diagnosis is based on the symp<strong>to</strong>ma<strong>to</strong>logy <strong>and</strong> priorcontact with a likely source of infection. Labora<strong>to</strong>ry confirmation is based on: (a)isolation of the etiologic agent from the patient’s local lesion, lymph nodes, <strong>and</strong>sputum by means of direct culture or inoculation in<strong>to</strong> labora<strong>to</strong>ry animals; (b) theimmunofluorescence test on exudates, sputum, <strong>and</strong> other contaminated materials;(c) the skin test with bacterial allergen, which gives delayed hypersensitivity reactions(these reagents can give a diagnosis during the first week of illness); <strong>and</strong> (d)serologic tests, such as tube agglutination or microagglutination (Snyder, 1980;Sa<strong>to</strong> et al., 1990). An enzyme-linked immunosorbent assay with sonicated antigenhas been perfected (Viljanen et al., 1983). This test has the advantage of permittingan early diagnosis, which is important for treatment; it can also detect IgM, IgA, orIgG antibodies. However, the microagglutination test is used more often due <strong>to</strong> itssimplicity <strong>and</strong> reliability; other tests are used only in case of doubt (Syrjälä et al.,1986). In the agglutination test, a four-fold increase in titer is significant.Antibodies appear in the second week of illness <strong>and</strong> may persist for years. Crossagglutination with Brucella antigen can occur, but at a lower level than with thehomologous antigen. Absorption of the patient’s serum with Brucella antigenremoves all doubt.In sheep, labora<strong>to</strong>ry confirmation is obtained by isolating the causal agent or byserologic tests.Due <strong>to</strong> the risk <strong>to</strong> labora<strong>to</strong>ry personnel, methods <strong>to</strong> isolate the causal agent shouldonly be used at reference labora<strong>to</strong>ries that have the required safety measures.Control: To prevent the disease in man, general <strong>and</strong> individual protective measuresmay be taken. General measures include reducing the source of infection, controllingvec<strong>to</strong>rs, changing the environment, <strong>and</strong> educating the public. Except for thelast one, these control measures are costly <strong>and</strong> difficult <strong>to</strong> apply. In the former Soviet


TULAREMIA 281Union, where tularemia was an important health problem, anti-tularemia instituteshave been established in epizootic regions <strong>to</strong> carry out these control activities.An important protective measure consists of immunizing at-risk individuals, populations,or occupational groups with attenuated live vaccines. In the former SovietUnion, the drastic reduction achieved in human morbidity is attributed <strong>to</strong> this singleactivity. In the US, there is an attenuated vaccine for high-risk groups (Burke, 1977)that has proven effective in reducing the incidence of the typhoidal form <strong>and</strong> attenuatingthe ulcerogl<strong>and</strong>ular form (Rohrbach, 1988). Other protective measures consis<strong>to</strong>f using insect repellents <strong>and</strong> protective clothing <strong>to</strong> avoid tick infestation <strong>and</strong>bites of other arthropods, promptly removing ticks from the body, using gloves <strong>to</strong>h<strong>and</strong>le <strong>and</strong> skin wild animals, avoiding consumption of untreated water in areaswhere contamination by F. tularensis is suspected, <strong>and</strong> thoroughly cooking wild animalmeat in enzootic areas.Controlling the infection in sheep involves applying tickicides by spray or dip,<strong>and</strong> administering antibiotics (strep<strong>to</strong>mycin, tetracyclines) in case of an outbreak.BibliographyAkerman, M.B., J.A. Embil. Antibodies <strong>to</strong> Francisella tularensis in the snowshoe hare(Lepus americanus struthopus) populations of Nova Scotia <strong>and</strong> Prince Edward Isl<strong>and</strong> <strong>and</strong> inthe moose (Alces alces americana Clin<strong>to</strong>n) population of Nova Scotia. Can J Microbiol28:403–405, 1982.Arata, A., H. Chamsa, A. Farhang-Azad, O. Mescerjakova, V. Neronov, S. Saidi. First detectionof tularaemia in domestic <strong>and</strong> wild mammals in Iran. Bull World Health Organ49:597–603, 1973.Baldwin, C.J., R.J. Panciera, R.J. Mor<strong>to</strong>n, et al. Acute tularemia in three domestic cats. JAm Vet Med Assoc 199:1602–1605, 1991.Bell, J.F, J.R. Reilly. Tularemia. In:Davis, J.W., L.H. Karstad, D.O. Trainer, eds. InfectiousDiseases of Wild Mammals. 2nd ed. Ames: Iowa State University Press; 1981.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Boyce, J.M. Francisella tularensis (Tularemia). In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E.Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: ChurchillLivings<strong>to</strong>ne, Inc.; 1990.Burke, D.S. Immunization against tularemia: Analysis of the effectiveness of liveFrancisella tularensis vaccine in prevention of labora<strong>to</strong>ry-acquired tularemia. J Infect Dis135:55–60, 1977.Capellan, J., I.W. Fong. Tularemia from a cat bite: Case report <strong>and</strong> review of feline-associatedtularemia. Clin Infect Dis 16:472–475, 1993.Frank, F.W., W.A. Meinershagen. Tularemia epizootic in sheep. Vet Med 56:374–378, 1961.Gelman, A.C. Tularemia. In: May, J.M., ed. Studies in Disease Ecology. New York:Hafner; 1961.Hornick, R.B. Tularemia. In: Hoeprich, P.D., ed. Infectious Diseases. Hagers<strong>to</strong>wn: Harper& Row; 1972.Marsh, H. Newsom’s Sheep Diseases. 2nd ed. Baltimore: Williams & Wilkins; 1958.McKeever, S., J.H. Schubert, M.D. Moody, et al. Natural ocurrence of tularemia in marsupials,carnivores, lagomorphs, <strong>and</strong> large rodents in southwestern Georgia <strong>and</strong> northwesternFlorida. J Infect Dis 103:120–126, 1958.Meyer, K.F. Pasteurella <strong>and</strong> Francisella. In: Dubos, R.J., J.G. Hirsch, eds. Bacterial <strong>and</strong>Mycotic Infections of Man. 4th ed. Philadelphia: Lippincott; 1965.


282 BACTERIOSESMörner, T., G. S<strong>and</strong>ström, R. Mattsson, P.O. Nilsson. Infections with Francisella tularensisbiovar palaearctica in hares (Lepus timidus, Lepus europaeus) from Sweden. J Wildl Dis24:422–433, 1988.Nayar, G.P., G.J. Crawshaw, J.L. Neufeld. Tularemia in a group of nonhuman primates. JAm Vet Med Assoc 175:962–963, 1979.Olsen, P.F. Tularemia. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger, eds.Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.Olsufjev, N.G., I.S. Meshcheryakova. Subspecific taxonomy of Francisella tularensis,McCoy <strong>and</strong> Chapin, 1912. Int J Syst Bacteriol 33:872–874, 1983.Pavlosky, E.N. Natural Nidality of Transmissible Diseases. Urbana: University of IllinoisPress; 1966.Reilly, J.R. Tularemia. In:Davis, J.W., L. Karstad, D.O. Trainer, eds. Infectious Diseases ofWild Mammals. Ames: Iowa State University Press; 1970.Rohrbach, B.W. Tularemia. J Am Vet Med Assoc 193:428–432, 1988.S<strong>and</strong>ström, G., A. Sjöstedt, M. Forsman, et al. Characterization <strong>and</strong> classification of strainsof Francisella tularensis isolated in the central Asian focus of the Soviet Union <strong>and</strong> in Japan.J Clin Microbiol 30:172–175, 1992.Sa<strong>to</strong>, T., H. Fujita, Y. Ohara, M. Homma. Microagglutination test for early <strong>and</strong> specificserodiagnosis of tularemia. J Clin Microbiol 28:2372–2374, 1990.Scheel, O., T. Hoel, T. S<strong>and</strong>vik, B.P. Berdal. Susceptibility pattern of Sc<strong>and</strong>inavianFrancisella tularensis isolates with regard <strong>to</strong> oral <strong>and</strong> parenteral antimicrobial agents. APMIS101:33–36, 1993.Schmid, G.P., A.N. Kornblatt, C.A. Connors, et al. Clinically mild tularemia associatedwith tick-borne Francisella tularensis. J Infect Dis 148:63–67, 1983.Snyder, M.J. Immune response <strong>to</strong> Francisella. In: Rose, H.R., H. Friedman, eds. Manual ofClinical Immunology. 2nd ed. Washing<strong>to</strong>n, D.C.: American Society for Microbiology; 1980.Syrjälä, H., P. Koskela, T. Ripatti, et al. Agglutination <strong>and</strong> ELISA methods in the diagnosisof tularemia in different clinical forms <strong>and</strong> severities of the disease. J Infect Dis153:142–145, 1986.Taylor, J.P., G.R. Istre, T.C. McChesney, et al. Epidemiologic characteristics of humantularemia in the southwest-central states, 1981–1987. Am J Epidemiol 133:1032–1038, 1991.Thorpe, B.D., R.W. Sidwell, D.E. Johnson, K.L. Smart, D.D. Parker. Tularemia in thewildlife <strong>and</strong> lives<strong>to</strong>ck of the Great Salt Lake Desert Region, 1951 through 1964. Am J TropMed Hyg 14:622–637, 1965.Tiggert, W.D. Soviet viable Pasteurella tularensis vaccines. A review of selected articles.Bacteriol Rev 26:354–373, 1962.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Tularemia associated with domestic cats—Georgia, NewMexico. MMWR Morb Mortal Wkly Rep 31:39–41, 1982.Viljanen, M.K., T. Nurmi, A. Salminen. Enzyme-linked immunosorbent assay (ELISA)with bacterial sonicate antigen for IgM, IgA, <strong>and</strong> IgG antibodies <strong>to</strong> Francisella tularensis:Comparison with bacterial agglutination test <strong>and</strong> ELISA with lipopolysaccharide antigen. JInfect Dis 148:715–720, 1983.Woodward, T.E. Tularemia. In: Beeson, P.B., W. McDermott, eds. Cecil Textbook ofMedicine. 15th ed. Philadelphia: Saunders; 1979.Zidon, J. Tularemia. In:Van der Hoeden, J., ed. Zoonoses. Amsterdam: Elsevier; 1964.


ZOONOTIC TUBERCULOSIS 283ZOONOTIC TUBERCULOSISICD-10 A16 respira<strong>to</strong>ry tuberculosis, not confirmedbacteriologically or his<strong>to</strong>logically; A18 tuberculosis of other organsEtiology: The etiologic agents of mammalian tuberculosis are Mycobacteriumtuberculosis, the main cause of human tuberculosis; M. bovis, the agent of bovinetuberculosis; <strong>and</strong> M. africanum,which causes human tuberculosis in tropical Africa.This last species has characteristics halfway between those of M. tuberculosis <strong>and</strong>M. bovis. M. microti,which causes tuberculosis in rodents, should be added <strong>to</strong> theseagents, although it is not of zoonotic interest (nontuberculous mycobacteria are presentedin the chapter, “Diseases Caused by Nontuberculous Mycobacteria”).The principal agent of zoonotic tuberculosis is M. bovis; the agent in man <strong>and</strong>other primates is M. tuberculosis, which is the type species of the genus.Tuberculous mycobacteria are alcohol- <strong>and</strong> acid-resistant, nonsporogenic, grampositivebacilli. These mycobacteria are resistant <strong>to</strong> many disinfectants, desiccation,<strong>and</strong> other adverse environmental fac<strong>to</strong>rs because the cell wall has a high lipidcontent.Phage typing is being used in epidemiological research on M. tuberculosis, <strong>and</strong>the API ZIM system divides the genus in<strong>to</strong> seven biovars (Casal <strong>and</strong> Linares, 1985;Humble et al., 1977). The use of phage typing was not widespread <strong>and</strong> the methodpractically fell in<strong>to</strong> disuse. It has been replaced by DNA hybridization.Analysis of DNA fragments obtained through the digestive action of one or morerestriction endonucleases is useful for identifying strains of both M. tuberculosis <strong>and</strong>M. bovis (Collins <strong>and</strong> De Lisle, 1985; Shoemaker et al., 1986).Many authors prefer <strong>to</strong> refer <strong>to</strong> a single species (M. tuberculosis) <strong>and</strong> human <strong>and</strong>bovine types.Geographic Distribution: The distribution of M. bovis <strong>and</strong> M. tuberculosis isworldwide. M. africanum is prevalent in Africa, but it has also been isolated inGermany <strong>and</strong> Engl<strong>and</strong>. M. africanum strains phenotypically related <strong>to</strong> M. tuberculosisare nitrase positive <strong>and</strong> are found in western Africa; those that are similar <strong>to</strong> M.bovis are nitrase negative <strong>and</strong> are isolated more frequently in eastern Africa (Grange<strong>and</strong> Yates, 1989).Occurrence in Man: The prevalence of human tuberculosis of animal origin hasdiminished greatly in countries where m<strong>and</strong>a<strong>to</strong>ry pasteurization of milk has beenimplemented <strong>and</strong> where successful campaigns <strong>to</strong> control <strong>and</strong> eradicate the bovineinfection have been carried out. The British Isles, where the incidence of humaninfection due <strong>to</strong> M. bovis is currently low <strong>and</strong> is limited <strong>to</strong> the elderly, were once themost affected area due <strong>to</strong> the consumption of raw milk. However, despite the greatreduction in rates of human infection by bovine strains in Great Britain, tuberculosisoriginated by these strains continues <strong>to</strong> occur. From 1977 <strong>to</strong> 1979 in southeastEngl<strong>and</strong>, isolations from 5,021 tuberculosis patients revealed 63 patients (1.25%)infected with “classic bovine strains” (M. bovis), 53 of which were Europeans <strong>and</strong>10, immigrants. Of these cases, 27 (42.85%) had pulmonary tuberculosis <strong>and</strong> 36(57.14%) had extrapulmonary tuberculosis. There was a marked difference in thefrequency of renal tuberculosis caused by M. bovis (23.8%) <strong>and</strong> M. tuberculosis


284 BACTERIOSES(8.2%). Commenting on these results, Collins et al. (1981) suggested the possibilityof human-<strong>to</strong>-human transmission, given that bovine tuberculosis had practically disappearedfrom Great Britain, that milk is pasteurized, <strong>and</strong> that some cases occurredin young people. Also in southeast Engl<strong>and</strong>, human cases caused by M. bovis continued<strong>to</strong> occur nearly 30 years after the program <strong>to</strong> eradicate bovine tuberculosisended in 1960. From 1977 <strong>to</strong> 1987, there were 201 new confirmed human casescaused by M. bovis, or 1.20% of all isolations of tuberculous mycobacteria (Yates<strong>and</strong> Grange, 1988). Most cases occurred in the elderly, who may have acquired theinfection when it was still prevalent in cattle (in 1935, before the start of the eradicationprogram, 40% of cattle were positive <strong>to</strong> tuberculin). The pulmonary <strong>and</strong> geni<strong>to</strong>urinaryforms are currently the most <strong>common</strong> forms in humans infected by M.bovis (Yates <strong>and</strong> Grange, 1988). In Slovakia, 52 human cases caused by the bovinebacillus were recorded during the period 1979–1983, 10 <strong>to</strong> 15 years after the eradicationof bovine tuberculosis. The average age of the patients was 61. Of these, 88%suffered from pulmonary tuberculosis; 17% of these were relapses <strong>and</strong> 71% werenew cases (Burjanova <strong>and</strong> Nagyova, 1985). In the Czech Republic, 47 patientsinfected by M. bovis were reported during the period 1981–1983 (Kubin et al.,1985). In Germany during the period 1953–1957, when the prevalence of tuberculosisin cattle was still high, 45% of tuberculous adenitis cases in children werecaused by M. bovis. Later, as prevalence declined in cattle, this form of tuberculosisas well as cutaneous tuberculosis declined notably. In the US at the beginning of the20th century, up <strong>to</strong> 20% of human tuberculosis was attributed <strong>to</strong> M. bovis; in 1980,barely 0.1% of human tuberculosis was so attributed (Good <strong>and</strong> Snider, 1980). In theNetherl<strong>and</strong>s, where bovine tuberculosis had been eradicated, 125 people wereinfected by M. bovis from 1972 <strong>to</strong> 1975 (Schonfeld, 1982). More than 80% of thesepatients were born when transmission of M. bovis via milk was still possible. Thefive patients younger than 20 years, who were born after the bovine infection waseradicated, were presumed <strong>to</strong> have contracted the infection outside the Netherl<strong>and</strong>s.Interhuman transmission is still a matter of controversy, but it is undeniable thateradication campaigns against bovine tuberculosis have drastically reduced the incidenceof human cases of this origin. For example, in Great Britain in 1945, 5% ofall fatal tuberculosis cases <strong>and</strong> 30% of cases of the disease in children under 5 yearsold were due <strong>to</strong> bovine strains (Collins <strong>and</strong> Grange, 1983).In countries where milk is routinely boiled, as in Latin America, the incidence ofinfection by M. bovis has always been low. Nevertheless, pulmonary <strong>and</strong> extrapulmonaryforms of human tuberculosis of animal origin continue <strong>to</strong> be a problem inareas where the prevalence of infection in cattle is high, because not all milk consumedis boiled, many products are prepared from unpasteurized milk, <strong>and</strong> cases ofinfection are contracted via aerosols. In Peru, a study of 853 strains of pulmonarytuberculosis identified 38 (4.45%) as M. bovis (Fernández Salazar et al., 1983).Several labora<strong>to</strong>ries in Argentina studied a <strong>to</strong>tal of 7,195 strains, primarily between1978 <strong>and</strong> 1981. Most of the strains were isolated from adult pulmonary tuberculosispatients, <strong>and</strong> 82 (1.1%) were classified as M. bovis (Argentina, ComisiónNacional de Zoonosis, 1982).Occurrence in Animals: In industrialized countries, bovine tuberculosis has beeneradicated or is in an advanced stage of control, while in several developing countriesthe situation has not improved or prevalence is increasing. Almost all Western


ZOONOTIC TUBERCULOSIS 285European countries report a prevalence of bovine infection lower than 0.1%. In theWestern Hemisphere, Canada <strong>and</strong> the US have reduced the infection rate <strong>to</strong> very lowlevels. In the US in 1969, 0.06% of 4.5 million cattle examined reacted <strong>to</strong> tuberculin(most of the reac<strong>to</strong>rs showed no evident lesions when slaughtered). In 1989, 33.5million cattle were slaughtered in the US (excluding reac<strong>to</strong>rs <strong>to</strong> tuberculin), ofwhich only 143 had tuberculous lesions (0.0004%). In Latin America, Costa Rica,Cuba, Jamaica, Panama, Uruguay, <strong>and</strong> Venezuela have national control programs.Cuba is already in the post-eradication surveillance phase. The rate of infection isvery low in several Central American <strong>and</strong> Caribbean countries. The highest infectionrates are found in the milk-producing regions near large cities in South America.In South American countries in which hogs are fed unpasteurized milk products, theinfection rate in swine is similar <strong>to</strong> or higher than in cattle, judging by records ofconfiscations at slaughterhouses. However, it should be kept in mind that these figuresinclude a large percentage of lesions caused by nontuberculous mycobacteria(see the chapter, “Diseases Caused by Nontuberculous Mycobacteria”).Bovine tuberculosis is important not only because it is a source of human infection,but also because of the economic losses it causes.Mycobacterium africanum, isolated for the first time from a human patient inSenegal <strong>and</strong> described in 1969 (Castets et al., 1969), is capable of infecting nonhumanprimates <strong>and</strong> causing pulmonary, lymph node, <strong>and</strong> renal lesions. Thorel (1980)isolated these strains from chimpanzees <strong>and</strong> from a Cercopithexus monkey ofAfrican origin that were found in experimental stations in Europe. These animalshad probably contracted the infection from man. There is a potential danger ofretransmission <strong>to</strong> those who work with them. There is also a report on bovine infectionin Malawi caused by M. africanum (Berggren, 1981), but it fails <strong>to</strong> providedetails on typing (Pritchard, 1988).Man can transmit M. tuberculosis <strong>to</strong> monkeys, dogs, cats, <strong>and</strong> psittacine birds (seethe section on the disease in animals).The Disease in Man: M. bovis can cause the same clinical forms <strong>and</strong> pathologiclesions as M. tuberculosis (the agent of human tuberculosis). His<strong>to</strong>rically, the mostprevalent forms caused by M. bovis were extrapulmonary, <strong>and</strong> children were amongthose most affected. The reason for extrapulmonary localization of the bovine bacillusis not that it has an affinity for other tissues, but that it is most <strong>common</strong>ly transmittedby consumption of raw milk or raw milk products. Thus, in those countrieswhere the prevalence of bovine tuberculosis was high <strong>and</strong> raw milk was consumed,many cases of extrapulmonary tuberculosis, such as cervical adenitis, geni<strong>to</strong>urinaryinfections, tuberculosis of the bones <strong>and</strong> joints, <strong>and</strong> meningitis, were caused by M.bovis. According <strong>to</strong> data on typing of tuberculosis bacilli in the British Isles prior <strong>to</strong>control of bovine infection, 50% or more of cervical adenitis cases were caused byM. bovis. Pulmonary tuberculosis caused by the bovine bacillus occurs less frequently,but its incidence is significant in occupational groups in contact withinfected cattle or their carcasses, particularly in countries where animals are stabled.This form cannot be distinguished clinically or radiologically from the diseasecaused by M. tuberculosis. Transmission occurs by aerosol droplets micromillimetersin diameter. In countries where the incidence of the human infection caused byM. tuberculosis has declined <strong>and</strong> the bovine infection has not been controlled, it isbelieved that M. bovis could assume a principal role in human pulmonary tubercu-


286 BACTERIOSESlosis. Although Denmark was declared free of bovine tuberculosis in 1952, 127cases of human infection caused by M. bovis, 58% of which were pulmonary tuberculosis,were detected between 1959 <strong>and</strong> 1963 in middle-aged <strong>and</strong> elderly persons.In countries where the control of bovine tuberculosis is advanced, human casescaused by M. bovis are observed mainly in the elderly, who were exposed <strong>to</strong> thepathogenic agent in their youth or childhood.Reduction or elimination of M. bovis in cattle <strong>and</strong> compulsory pasteurization ofmilk have helped reduce the incidence of infection in man. At the same time, the clinicalpicture of human infection caused by the bovine agent has changed. Currently,pulmonary tuberculosis predominates, followed by urogenital tuberculosis.Interhuman transmission of M. bovis is possible, but few cases have been satisfac<strong>to</strong>rilyconfirmed. As is the case with most <strong>zoonoses</strong>, man is generally an accidentalhost of M. bovis <strong>and</strong> human infection depends on the animal source. Since M. tuberculosis<strong>and</strong> M. bovis are very similar in their pathogenic effect on man, it is not unders<strong>to</strong>odwhy large-scale interhuman transmission of the bovine infection does not occur.A possible explanation is that pulmonary patients infected by M. bovis shed fewer bacteriain their sputum than do those infected by M. tuberculosis (Griffith, 1937).Inhabitants of Latin America have been assumed <strong>to</strong> be protected from infectionby the bovine bacillus because of the widespread cus<strong>to</strong>m of boiling milk.Undoubtedly, if this practice were not followed, the rate of human infection by M.bovis would be much higher there, considering the infection’s wide distribution <strong>and</strong>the rate of infection in dairy cattle in many Latin American countries. However,some people in rural areas do drink raw milk <strong>and</strong> frequently consume products(cream, butter, soft cheese) made at home from raw milk. In Latin America <strong>and</strong> otherparts of the world, children are the main victims, as indicated by typing data fromBrazil, Peru, <strong>and</strong> Mexico. These data also confirm that some children are fed milkor milk products that are not heat-treated.Although it is not cus<strong>to</strong>mary <strong>to</strong> stable cattle in Latin America, cases of pulmonarytuberculosis caused by M. bovis have been recorded, with rural laborers <strong>and</strong> employeesof abat<strong>to</strong>irs <strong>and</strong> locker plants being the most exposed groups. In Argentina, thebovine bacillus was isolated from 8% of 85 pulmonary patients from rural areas,while only 1 case due <strong>to</strong> M. bovis was found among 55 patients in the capital.People suffering from pulmonary tuberculosis of bovine origin can, in turn,retransmit the infection <strong>to</strong> cattle. This occurrence is particularly evident in herdsfrom which tuberculosis has been eradicated <strong>and</strong> which later become reinfected, thesource of exposure often being a ranch h<strong>and</strong> with M. bovis tuberculosis. Suchepisodes have occurred in the US <strong>and</strong> in several European countries. Between 1943<strong>and</strong> 1952, 128 herds containing more than 1,000 head of cattle were reinfected inDenmark by 107 individuals with tuberculosis. Similar occurrences continued inDenmark until 1960, despite advances made in the eradication of bovine tuberculosis.Huitema (1969) reports on 50 herds that were infected by people with tuberculosiscaused by M. bovis; 24 of the patients suffered from renal tuberculosis. It ispossible that this phenomenon (retransmission of the infection from man <strong>to</strong> cattle)also occurs in the Southern Hemisphere, but goes unnoticed due <strong>to</strong> high rates oftuberculosis in cattle.In regions where bovine tuberculosis has been eradicated, cattle cease <strong>to</strong> be asource for human infection, but man may continue <strong>to</strong> be a potential source of infectionfor cattle for years.


ZOONOTIC TUBERCULOSIS 287Persons with pulmonary or geni<strong>to</strong>urinary tuberculosis due <strong>to</strong> the human typespecies (M. tuberculosis) can temporarily infect <strong>and</strong> sensitize cattle. Cattle are veryresistant <strong>to</strong> M. tuberculosis; the agent does not cause a progressive tuberculosis inthese animals, but the bacillus can survive for some time in their tissues, especiallythe lymph nodes, sensitizing the animal <strong>to</strong> mammalian tuberculin <strong>and</strong> confusing thediagnosis. Sensitization can persist for some six <strong>to</strong> eight months after the humansource of infection is removed. Elimination of M. tuberculosis in milk has occasionallybeen confirmed, but tuberculous lesions of the udder were not present. Mancan transmit the human bacillus <strong>to</strong> several other animals, principally monkeys <strong>and</strong>dogs, in which it produces a progressive tuberculosis.In many countries, direct or indirect exposure of man <strong>to</strong> bovine tuberculosis is animportant source of sensitization <strong>to</strong> tuberculin. In Denmark, a relationship wasfound between the prevalence of bovine tuberculosis <strong>and</strong> the rate of reac<strong>to</strong>rs <strong>to</strong>tuberculin in the human population. In the same country, statistical data indicate thata third of the population between the ages of 30 <strong>and</strong> 35 owes its tuberculin sensitization<strong>to</strong> infection by M. bovis. The same study suggests that the risk of developingpulmonary tuberculosis later is much smaller among those sensitized by the bovinebacillus than by the human bacillus, perhaps because M. bovis infection is contractedmainly through the digestive tract <strong>and</strong> not via aerosols. Another interestingconclusion is that less calcification occurs in pulmonary tuberculosis resulting fromM. bovis than from M. tuberculosis.The treatment for humans infected by M. bovis is the same as for those infectedby M. tuberculosis (isoniazid, rifampicin, ethambu<strong>to</strong>l), except that pyrazinamideshould be excluded, as it is not active against the bovine bacillus.The Disease in Animals: Many mammalian species are susceptible <strong>to</strong> the agentsof tuberculosis. Bovine tuberculosis is the most important form in economic terms<strong>and</strong> as a zoonosis. Tuberculosis in swine also causes substantial economic losses.CATTLE: The principal etiologic agent for cattle is M. bovis. As in man, the bacillusenters the body mainly by inhalation. The intestinal tract is an important route ofinfection in calves nursed on contaminated milk. The most <strong>common</strong> clinical <strong>and</strong>pathological form is pulmonary tuberculosis. The causal agent enters the lungs <strong>and</strong>multiplies there, forming the primary focus; this is accompanied by tuberculouslesions in the bronchial lymph nodes of the same side, thus producing the primarycomplex. These lesions can remain latent or develop further, depending on the interactionbetween the agent <strong>and</strong> the host’s body. If the animal’s resistance <strong>to</strong> tuberculosisbacilli breaks down, the infection will spread <strong>to</strong> other organs via the lymph orblood vessels, giving rise <strong>to</strong> early generalization of the infection. If the immune systemis unable <strong>to</strong> destroy the bacilli, they will cause tubercles <strong>to</strong> form in organs <strong>and</strong>tissues where they lodge. New foci are produced, mainly in the lungs, kidneys, liver,spleen, <strong>and</strong> their associated lymph nodes. Dissemination may also give rise <strong>to</strong> acutemiliary tuberculosis.In most cases, tuberculosis has a chronic course, with effects limited <strong>to</strong> the lungs. Thedisease process is slow <strong>and</strong> may remain clinically inapparent for a long time. In fact,some animals spend their entire useful lives without any evident symp<strong>to</strong>ma<strong>to</strong>logy,although they constitute a potential threat for the rest of the herd. Other animals developchronic bronchopneumonia, accompanied by coughing <strong>and</strong> reduced milk production.In advanced cases, when the lungs are largely destroyed, there is pronounced dyspnea.


288 BACTERIOSESPearl disease, a tuberculous peri<strong>to</strong>nitis or pleurisy, is another form sometimesobserved in infected herds in countries with no tuberculosis control program.It is estimated that about 5% of tuberculous cows, especially in advanced cases,have tuberculous uterine lesions or tuberculous metritus, <strong>and</strong> that 1% <strong>to</strong> 2% havetuberculous mastitis. This clinical form not only has public health repercussions, butalso serves as a source of infection for calves nursed naturally or artificially. One ofthe main signs of tuberculosis acquired by the oral route is swelling of the retropharyngeallymph nodes. In calves, the primary lesion is usually located in the mesentericlymph nodes <strong>and</strong> the intestinal mucosa is not affected.The disease appears more frequently in older animals because the disease ischronic <strong>and</strong> because older animals have had more time <strong>to</strong> be exposed <strong>to</strong> the infection.The infection is more prevalent among dairy cattle than among beef cattle,not only because their useful economic life is longer, but because dairy cattle arein closer contact with one another when gathered for milking or when housed indairy sheds.Cattle are resistant <strong>to</strong> the M. avium complex (MAC) <strong>and</strong> rarely suffer progressivetuberculosis due <strong>to</strong> these agents. Nevertheless, they are very important in controlprograms because cattle can become paraspecifically sensitized <strong>to</strong> mammaliantuberculin, leading <strong>to</strong> difficulties in diagnosis. M. avium infects cattle through thedigestive tract. When lesions are present, they are generally limited <strong>to</strong> the intestine<strong>and</strong> mesenteric lymph nodes. However, lesions can occasionally be found in thelungs <strong>and</strong> regional lymph nodes but not in other tissues, indicating that the entryroute may sometimes be the respira<strong>to</strong>ry tract. Lesions tend <strong>to</strong> heal spontaneously.Bovine-<strong>to</strong>-bovine transmission of M. avium infection does not occur (see the chapter,“Diseases Caused by Nontuberculous Mycobacteria”).Cattle are very resistant <strong>to</strong> M. tuberculosis, <strong>and</strong> rarely develop ana<strong>to</strong>micopathologiclesions. In several countries, M. tuberculosis has been isolated from the lymphnodes of some positive reac<strong>to</strong>rs <strong>to</strong> tuberculin that showed no lesions in postmortemexamination. Again in this instance, the infection’s importance lies in sensitizingthese animals <strong>to</strong> tuberculin.An experiment comparing the pathogenicity of M. africanum, M. bovis, <strong>and</strong> M.tuberculosis for calves inoculated intravenously showed that M. africanum (at leastthe strain used in this experiment) was as pathogenic for calves as M. bovis (deKan<strong>to</strong>r et al., 1979).SWINE: This species is susceptible <strong>to</strong> the following agents: M. bovis, M. aviumcomplex, <strong>and</strong> M. tuberculosis. M. bovis is the most pathogenic <strong>and</strong> invasive forswine <strong>and</strong> is the cause of most cases of generalized tuberculosis.The principal route of infection is the digestive tract through consumption of contaminatedmilk or milk products, kitchen <strong>and</strong> abat<strong>to</strong>ir scraps, <strong>and</strong> excreta fromtuberculous fowl <strong>and</strong> cattle. The primary infection complex is found in the oropharynx<strong>and</strong> the submaxillary lymph nodes, or in the intestines <strong>and</strong> the mesenteric lymphnodes. The lesions are usually confined in the primary complex. Chronic lesions arenot found in single organs, as they often are in cattle. Prevalence is lower in younganimals than in adults, but the former show a greater tendency <strong>to</strong>ward generalizationof the infection. Eradication programs for bovine tuberculosis directly help <strong>to</strong>reduce the infection rate among swine. In the US in 1924, tuberculous lesions werefound in 15.2% of hogs butchered, while in 1989, they were found in only 0.67%.


ZOONOTIC TUBERCULOSIS 289Most cases of swine tuberculosis are due <strong>to</strong> the M. avium complex. Thus, the reductionof avian tuberculosis has also helped lower the rate of infection in swine. InGreat Britain, as tuberculosis of bovine origin declined, infections caused by MACincreased proportionally (Lesslie et al., 1968). The <strong>to</strong>tal number of confiscationsdue <strong>to</strong> generalized tuberculosis was reduced even more drastically. In some LatinAmerican countries, M. bovis is the cause of 80% <strong>to</strong> 90% of tuberculous lesions inswine. The relative proportions of M. bovis <strong>and</strong> MAC as the cause of swine tuberculosisare reversed when M. bovis infection is controlled in cattle, as it has been inseveral European countries <strong>and</strong> in the US.MAC usually causes adenitis of the digestive tract <strong>and</strong>, more rarely, a generalizeddisease (see the chapter, “Diseases Caused by Nontuberculous Mycobacteria”).Swine are also susceptible <strong>to</strong> the human bacillus (M. tuberculosis), which producesan infection of the lymph nodes that drain the digestive system <strong>and</strong>, morerarely, generalized tuberculosis. The main sources of infection are kitchen scraps<strong>and</strong> lef<strong>to</strong>vers from tuberculosis sana<strong>to</strong>riums. This infection has been confirmed inseveral countries in the Americas, Europe, <strong>and</strong> Africa.Swine-<strong>to</strong>-swine transmission of the infection is insignificant. Intestinal lesions arehyperplastic, <strong>and</strong> ulcers that would cause the agent <strong>to</strong> be shed are not observed.However, swine may transmit the infection <strong>to</strong> other swine when they have pulmonary,uterine, or mammary lesions (Thoen, 1992).If there is generalization of the infection caused by MAC, the lesions appear indiffuse form <strong>and</strong> there is little tendency <strong>to</strong>ward encapsulation. The cutaway view ofa lesion generally shows a smooth surface <strong>and</strong> there may be foci of caseation, butcalcification is minimal. Lesions caused by M. bovis or M. tuberculosis, in contrast,are caseous <strong>and</strong> well-circumscribed by fibrosis with pronounced calcification(Thoen, 1992). Other bacteria, for example Rodococcus equi, can produce lesionssimilar <strong>to</strong> tuberculous lesions.SHEEP AND GOATS: Tuberculosis in sheep is generally rare <strong>and</strong> sporadic. In the fewcases described, the most important agent was M. avium,followed by M. bovis. Onlytwo cases involved M. tuberculosis. In research in New Zeal<strong>and</strong> stemming from aprogram <strong>to</strong> eradicate bovine tuberculosis, multiples cases of infection by M. boviswere confirmed among sheep sharing the same pasture with infected cattle. In onearea, 597 sheep were given the tuberculin test on the inner thigh <strong>and</strong> 108 (18%) reac<strong>to</strong>rswere discovered. Lesions, mostly in the lymph nodes, were found in 43 (61%)out of 70 necropsies. The lungs were affected in eight sheep (Davidson et al., 1981).A similar result was observed in another region of New Zeal<strong>and</strong>, on l<strong>and</strong> where theprevalence of tuberculosis in cattle <strong>and</strong> opossums (Trichosurus vulpecula) was high.The tuberculin test yielded positive results in 11% of the sheep, <strong>and</strong> was judged <strong>to</strong>have a sensitivity of 81.6% <strong>and</strong> a specificity of 99.6% (Cordes et al., 1981).Prevalence in goats seems <strong>to</strong> be low. In countries with advanced programs <strong>to</strong> eradicatebovine tuberculosis, the infection in goats is moni<strong>to</strong>red, since this species issusceptible <strong>to</strong> M. bovis, frequently suffers from pulmonary tuberculosis, <strong>and</strong> canreinfect cattle. Nannies also suffer from tuberculous mastitis <strong>and</strong> their milk mayconstitute a danger <strong>to</strong> the consumer. In addition, goats are susceptible <strong>to</strong> M. avium<strong>and</strong> M. tuberculosis, <strong>and</strong> the latter agent sometimes causes generalized processes.Little is known about the disease’s occurrence in goats in developing countries,since these animals are generally slaughtered without veterinary inspection.


290 BACTERIOSESHORSES: Tuberculosis is infrequent in horses. In countries where the incidence ofbovine infection is high, the principal agent of the disease in horses is M. bovis. Theinfection’s predominant route of entry is the digestive system. Lesions are generallyconfined <strong>to</strong> the lymph nodes of the digestive tract, where they produce a tissue reactionthat resembles tumors. Some cases of generalized infection, caused by both M.bovis <strong>and</strong> M. avium, have been described. Often, no lesions are found in infectionsproduced by M. avium. In Germany, the avian bacillus was isolated from 30% of 208horses with no apparent lesions.M. tuberculosis is seldom isolated from horses. In a study carried out sometime ago, only 13 of 241 typed strains corresponded <strong>to</strong> the human bacillus(Francis, 1958).The disease is very rare in asses <strong>and</strong> mules.It is interesting <strong>to</strong> note that horses are hypersensitive <strong>to</strong> tuberculin, <strong>and</strong> thus theallergenic test does not give reliable results.DOGS AND CATS: Dogs are resistant <strong>to</strong> experimental tuberculosis infection.Recorded cases in dogs are probably due <strong>to</strong> massive <strong>and</strong> repeated exposure broughtabout by living with humans with tuberculosis or frequently eating contaminatedfood. Infection may be produced by aerosols, or by ingestion of sputa, milk, <strong>and</strong> viscera.Almost 75% of the cases are due <strong>to</strong> the human bacillus <strong>and</strong> the rest <strong>to</strong> thebovine. The clinical picture is not characteristic. The only symp<strong>to</strong>ms found in eighttuberculous dogs in New York City were anorexia, weight loss, lethargy, vomiting,<strong>and</strong> leukocy<strong>to</strong>sis. Radiology revealed pleural <strong>and</strong> pericardial effusion, ascites, <strong>and</strong>hepa<strong>to</strong>megaly. Granuloma<strong>to</strong>us lesions in soft tissues were similar <strong>to</strong> those observedin neoplasias (Liu et al., 1980). Infection mainly localizes in the lungs or mesentericlymph nodes; intestinal ulcers <strong>and</strong> renal lesions are sometimes found as well.Consequently, dogs can shed bacilli by coughing <strong>and</strong> in their saliva, feces, <strong>and</strong> urine.It has also been demonstrated that the etiologic agent can be present in the pharynx<strong>and</strong> feces of dogs living in the same house with tuberculous patients, even when theanimals show no tuberculous lesions. Although few cases of transmission from dog<strong>to</strong> man have been confirmed, a tuberculous dog (or even an apparently healthy animalliving with a tuberculous patient) represents a potential risk <strong>and</strong> should bedestroyed. A dog infected with M. bovis can, in turn, be a potential source of reinfectionfor cattle.Cats also have a great natural resistance <strong>to</strong> tuberculosis. M. bovis is the most <strong>common</strong>pathogen in cats, <strong>and</strong> has been isolated in 90% of the cases. The agent gainsentry via the digestive tract when milk or viscera containing tuberculosis bacilli isconsumed. Cat-<strong>to</strong>-cat transmission of M. bovis in a scientific institution in Australiahas been described (Isaac et al., 1983). In countries where bovine tuberculosis hasbeen brought under control, infection in cats is rare, <strong>and</strong> the few recorded cases havebeen caused by M. tuberculosis <strong>and</strong> occasionally MAC.Destructive lesions are sometimes found; pneumonitis <strong>and</strong> cutaneous tuberculosisare frequent. In urban areas of Buenos Aires, a cooperative study was conducted bythe Pasteur Institute <strong>and</strong> the Pan American Institute for Food Protection <strong>and</strong>Zoonoses (INPPAZ). M. bovis was isolated from the lesions of 10 of approximately150 cats studied (I.N. de Kan<strong>to</strong>r. Personal communication). In New Zeal<strong>and</strong>between 1974 <strong>and</strong> 1986, M. bovis was isolated from 57 cats. With the exception ofsix animals, all came from suburban <strong>and</strong> rural areas where tuberculosis is also pres-


ZOONOTIC TUBERCULOSIS 291ent in wild animals, particularly the opossum (Trichosurus vulpecula). Cutaneouslesions were observed in 58% of the cats, with a pyogranuloma<strong>to</strong>us reaction <strong>and</strong>coagulative necrosis. These lesions had not been described earlier in other geographicareas, where the prevalence of tuberculosis among cats was 2% <strong>to</strong> 13%before successful control <strong>and</strong> eradication programs were implemented. Presumablythe cats acquired the infection when feeding on tuberculous wild animals (De Lisleet al., 1990). Several cases of reinfection of cattle herds by tuberculous cats havebeen described.WILD, CAPTIVE, AND DOMESTIC ANIMALS: Animals living in the wild, far from man<strong>and</strong> domestic animals, generally do not contract tuberculosis. On the other h<strong>and</strong>,captive animals in zoos, on pelt farms, in labora<strong>to</strong>ries, <strong>and</strong> in family homes may beexposed <strong>to</strong> infection. Monkeys are susceptible <strong>to</strong> M. tuberculosis as well as M.africanum <strong>and</strong> M. bovis. Almost 70% of the isolations from these animals are strainsof the human bacillus, some are M. africanum, <strong>and</strong> the rest are M. bovis. The diseaseis contracted via the respira<strong>to</strong>ry or digestive route. The infection can be propagatedfrom monkey <strong>to</strong> monkey <strong>and</strong> constitutes a grave problem for colonies kept inscientific institutions <strong>and</strong> zoos. These animals can retransmit the infection <strong>to</strong> man.It is not unusual <strong>to</strong> find tuberculous pet monkeys that may have been infected beforetheir acquisition or through contact with a family member. In France, infection due<strong>to</strong> M. africanum has been described in three chimpanzees <strong>and</strong> a Cercopithexus monkey.Three of these animals belonged <strong>to</strong> a scientific center <strong>and</strong> one of the chimpanzeesbelonged <strong>to</strong> a zoo. Since M. africanum has properties intermediate betweenthose of M. bovis <strong>and</strong> M. tuberculosis, it is possible that infection by M. africanumwas not described earlier in nonhuman primates because the species type of strainsisolated previously was misidentified. It still has not been determined whether theinfection was transmitted <strong>to</strong> the primates by man or acquired in their natural foresthabitat (Thorel, 1980).Tuberculosis is a problem in cervids, particularly now that deer farming hasbecome popular in several countries. Tuberculosis in deer is caused primarily by M.bovis. This presents the possibility of retransmission of the infection <strong>to</strong> cattle incountries that are practically free of bovine tuberculosis. It is also a potential risk forpeople who are in contact with these animals. M. bovis has been found in free-roamingdeer, probably living near cattle operations in Canada, the US (Hawaii), GreatBritain, Irel<strong>and</strong>, <strong>and</strong> Switzerl<strong>and</strong>. Deer most exposed <strong>to</strong> the disease are captive animalsin zoos or deer on farms.The first report on infection in farmed deer comes from New Zeal<strong>and</strong>, in a regionwhere the disease exists in cattle <strong>and</strong> opossum (Trichosurus vulpecula). An outbreakof bovine tuberculosis was recorded on farms in Engl<strong>and</strong> that imported red deer(Cervus elaphus) from an eastern European country. Upon necropsy of 106 deer, 26were found <strong>to</strong> be infected <strong>and</strong> 19 had visible lesions. The tuberculin test had 61.3%specificity <strong>and</strong> 80% sensitivity (Stuart et al. 1988). In a case of this type, the testwould be used primarily <strong>to</strong> determine whether or not tuberculosis exists on a farm.An eradication program has been established in New Zeal<strong>and</strong> based on the tuberculintest <strong>and</strong> slaughter of reac<strong>to</strong>rs, but the high percentage of false negatives hamperssuccess.In South Australia, an outbreak was reported in 1986 in three herds of anotherdeer species (Dama dama). Upon necropsy, 47 of 51 animals were found <strong>to</strong> have


292 BACTERIOSESbovine tuberculosis (Robinson et al., 1989). In eight US states, the infection wasfound in 1991 in ten herds of deer. This caused concern because information fromCanada indicated the possibility of human infection from this source (Essey et al.,1991).Outbreaks have been described in farmed fur-bearing animals, such as mink <strong>and</strong>silver fox; the source of infection was meat or viscera of tuberculous cattle or fowl.Tuberculosis has been found in wild species of ungulates <strong>and</strong> carnivores in zoos <strong>and</strong>some nature preserves, <strong>and</strong> the infection has been confirmed in several animalspecies in zoos in Latin America <strong>and</strong> other parts of the world.Two wild species are reservoirs of <strong>and</strong> sources of infection by M. bovis in cattle. Anopossum (Trichosurus vulpecula) from Australia—where tuberculous infection hasnot been found in this species—was introduced in<strong>to</strong> New Zeal<strong>and</strong>, where it contractedbovine tuberculosis. Currently, opossum are attributed a major role in maintaininginfection caused by M. bovis in cattle in the region of New Zeal<strong>and</strong> where they arefound. DNA restriction endonuclease analysis demonstrated that M. bovis isolatesfrom cattle <strong>and</strong> opossum in Upper Hutt (the region where the eradication programencountered difficulties) belong <strong>to</strong> the same restriction category (Collins et al., 1988).In southwestern Engl<strong>and</strong>, reinfection of cattle herds has been attributed <strong>to</strong> thehigh rate of M. bovis infection found in badgers (Meles meles). When the badgerpopulation was eliminated from certain areas <strong>and</strong> prevented from repopulatingthem, transmission <strong>to</strong> cattle was halted, thus proving the causal relationship betweeninfection in these species (Wilesmith, 1983).There is abundant literature on the infection in badgers <strong>and</strong> cattle. It is estimatedthat the badger population in Great Britain is approximately 250,000 animals <strong>and</strong>that infection by M. bovis is endemic on the isl<strong>and</strong>, regardless of the density of thecolonies. A <strong>to</strong>tal of 15,000 badgers, most of them killed on roadways, were examined<strong>and</strong> 3.9% were positive for M. bovis (Cheeseman et al., 1989). Theseresearchers <strong>and</strong> others (Wilesmith et al., 1986) consider the badger an ideal maintenancehost or natural reservoir that acts as a source of infection for cattle, althougha low level source. In Irel<strong>and</strong>, it was demonstrated that destroying badger coloniesreduced the prevalence of the disease in cattle. The highest incidence of tuberculosisin cattle was found in areas with a high population density of cattle <strong>and</strong> badgers(McAleer, 1990). Although these researchers admit that badgers may be partiallyresponsible for tuberculosis in cattle, there are other questions that must be investigated<strong>and</strong> clarified. As badgers scour pastures in search of worms, they excrete M.bovis in their feces, urine, <strong>and</strong> sputum <strong>and</strong> in pus when they have open abscesses. Itis not clear how the cattle become infected, as they suffer primarily from pulmonarytuberculosis, which has a respira<strong>to</strong>ry route of infection, except in the case of calves,many of which become infected by mouth through contaminated milk. Badgercolonies with tuberculosis may also coexist with cattle for some time without transmittingthe infection <strong>to</strong> these animals (Grange <strong>and</strong> Collins, 1987).In Argentina in 1982 <strong>and</strong> 1983, 4 million hares (Lepus europaeus) were slaughteredunder veterinary inspection <strong>and</strong> 369 animals were confiscated for variouscauses. M. bovis was isolated from only five hares <strong>and</strong> his<strong>to</strong>pathological examinationshowed a tuberculous granuloma with significant caseation <strong>and</strong> little calcification,with the presence of alcohol- <strong>and</strong> acid-resistant bacilli (de Kan<strong>to</strong>r et al., 1984).Alpacas imported from the Andean highl<strong>and</strong>s <strong>to</strong> Europe were the cause of small outbreaksof tuberculosis (Veen et al., 1991).


ZOONOTIC TUBERCULOSIS 293In South Africa, tuberculosis has been diagnosed in many wild species: the Capebuffalo (Syncerus caffer), the greater kudu (Tragelaphus strepsiceros), <strong>and</strong> the forestduiker (Cephalophus grimmia), among others (Pas<strong>to</strong>ret et al., 1988).Source of Infection <strong>and</strong> Mode of Transmission (Figure 20): The main reservoirof M. bovis is cattle, which can transmit the infection <strong>to</strong> many mammalian species,including man. Man contracts the infection primarily by ingesting the agent in rawmilk <strong>and</strong> milk products, <strong>and</strong> secondarily by inhaling it.Tuberculosis is transmitted among cattle mainly via aerosols. The digestive tractis an important route of transmission prior <strong>to</strong> weaning.A human infected by M. bovis who suffers from the pulmonary or urogenital formof tuberculosis can retransmit the infection <strong>to</strong> cattle. This phenomenon becomes particularlyevident during the final stages of bovine tuberculosis eradication.Tuberculosis in swine, goats, <strong>and</strong> sheep has as it principal source of infection cattle,fowl, <strong>and</strong> occasionally man. Swine are infected enterogenously, <strong>and</strong> retransmission<strong>to</strong> other swine, other species, <strong>and</strong> man is thought <strong>to</strong> be rare. Goats can constitutea source of infection for man <strong>and</strong> for cattle.Dogs often contract the infection from humans <strong>and</strong>, less frequently, from cattle.They may in turn retransmit it <strong>to</strong> man <strong>and</strong> cattle. Dogs become infected via thedigestive <strong>and</strong> respira<strong>to</strong>ry tracts. The principal source of infection for cats is cattle<strong>and</strong>, <strong>to</strong> a lesser degree, man. The route of entry is mainly oral. At times, cats can bea source of infection for cattle <strong>and</strong> humans.Among wild animals in captivity, monkeys are particularly interesting because oftheir susceptibility <strong>to</strong> M. tuberculosis <strong>and</strong> M. bovis. They contract the infection fromman by inhaling the agent. Tuberculous primates constitute a health risk for humans.Figure 20. Tuberculosis (Mycobacterium bovis). Mode of transmission.TuberculousbovineMainly airborne route;digestive route (suckling calves)BovineDigestive route, mainly throughraw milk <strong>and</strong> milk products;less frequently via airborne routeDigestive route, mainly through rawmilk <strong>and</strong> milk products;less frequently via airborne routeOccasionally reinfection of bovinesvia airborne routeSwine, sheep,goats, horses,dogs, cats,wild animalsMan


294 BACTERIOSESDomestic cattle are the source of infection for wild animals. Once the agent isintroduced among wild animals that share pasture with cattle, it can spread amongthem <strong>and</strong> represent a risk for domestic animals <strong>and</strong> for man. This is true of deer <strong>and</strong>badgers (Meles meles) in Great Britain <strong>and</strong> of opossum (Trichosurus vulpecula) inNew Zeal<strong>and</strong>.Role of Animals in the Epidemiology of the Disease: Human-<strong>to</strong>-human transmissionof animal tuberculosis is rare. The infection depends on an animal source.Diagnosis: Since the human infections caused by M. tuberculosis <strong>and</strong> M. bovisare clinically <strong>and</strong> radiologically indistinguishable, definitive diagnosis can only beachieved by isolating <strong>and</strong> typing the etiologic agent. In this regard, it should benoted that M. bovis grows poorly in media containing glycerin, such as Löwenstein-Jensen culture, which are generally used for culturing M. tuberculosis.For routine diagnosis of bovine tuberculosis, the only approved method for eradicationprograms is the tuberculin test. The most appropriate tuberculin is the purifiedprotein derivative (PPD), since it is specific <strong>and</strong> not very costly <strong>to</strong> produce. Ithas been made from both human <strong>and</strong> bovine strains, but research has shown thattuberculin produced with an M. bovis strain is more specific. In most countries, onlya PPD tuberculin is used in eradication campaigns, <strong>and</strong> the comparative test (simultaneousapplication of mammalian <strong>and</strong> avian tuberculin) is reserved for problemherds in which paraspecific sensitization is suspected. The test is carried out byintradermal inoculation of 0.1 ml of tuberculin in<strong>to</strong> the skin of the caudal fold or thewide part of the neck, depending on st<strong>and</strong>ards established in each country. It shouldbe borne in mind that the skin of the neck is much more sensitive than that of thecaudal fold. The amount of tuberculin used varies from 2,000 <strong>to</strong> 10,000 IU in differentcountries. The test will be more sensitive but less specific when larger dosesare used. The test’s effectiveness depends not only on the tuberculin <strong>and</strong> its correctapplication, but on the response capability of the infected animal. Some herdsinclude anergic animals, which are usually old <strong>and</strong> have very advanced tuberculosis.Clinical examination <strong>and</strong> knowledge of the herd’s his<strong>to</strong>ry can help <strong>to</strong> completethe diagnosis.The tuberculin test may also be applied <strong>to</strong> goats, sheep, <strong>and</strong> swine with satisfac<strong>to</strong>ryresults. In swine, the preferred inoculation site is the base of the ear, with 2,000IU of mammalian <strong>and</strong> avian tuberculin; in goats <strong>and</strong> sheep, the tuberculin can beapplied <strong>to</strong> the eyelid, the fold of the tail, or the inner thigh.The tuberculin test is unsatisfac<strong>to</strong>ry for horses, dogs, <strong>and</strong> cats. Some research hassuggested that the test using BCG might give better results in dogs. For monkeys,the intrapalpebral test is recommended, as well as radiography in advanced cases.The tuberculin test has several disadvantages. These include waiting time (readingat 72 hours in cattle) <strong>and</strong> the need for the veterinarian <strong>to</strong> visit the herd twice(once <strong>to</strong> inject the tuberculin <strong>and</strong> the other <strong>to</strong> read the test). Similarly, humanpatients require two medical visits. Old cows with advanced tuberculosis are anergic.This can also happen if there is an intercurrent febrile disease. As an eradicationprogram progresses, the percentage of reac<strong>to</strong>r animals without visible tuberculosislesions increases at slaughterhouses. These disadvantages have led many researchers<strong>to</strong> seek serologic tests than can replace or at least complement tuberculin tests.The enzyme-linked immunosorbent assay (ELISA) using bovine PPD was evaluatedin five different groups of cattle, including 53 animals with positive cultures


ZOONOTIC TUBERCULOSIS 295<strong>and</strong> 101 animals from a tuberculosis-free area. Sensitivity of 73.6% <strong>and</strong> specificityof 94.1% were obtained (Ritacco et al., 1990). The authors note that ELISA wasable <strong>to</strong> detect IgG against M. bovis in the sera of cattle with active tuberculosis, butnot in those with a clinically inapparent infection (e.g., at the onset of infection orin the latent state). There was little coincidence between the results from the tuberculintest <strong>and</strong> results from ELISA. Antibodies were detected in almost three out offour bovines with active tuberculosis. In contrast <strong>to</strong> what happens with anergic animals,which lose cellular reactivity <strong>to</strong> the hypersensitivity test with tuberculin, antibodiesare more abundant when there is a strong antigenic discharge. Thus, ELISAcould be useful as a complement <strong>to</strong> the intradermal test in detecting anergic tuberculousanimals that represent a risk for the rest of the herd (Ritacco et al., 1990). Theresults obtained in humans infected by M. tuberculosis are not unlike those obtainedin cattle infected by M. bovis. Specificity was 93% in adults <strong>and</strong> 98% in children;sensitivity was 69% in adults <strong>and</strong> 51% in children. The conclusion is that enzymeimmunoassay can be useful for detecting patients with nonbacilliferous, extrapulmonary,<strong>and</strong> pediatric tuberculosis (de Kan<strong>to</strong>r et al., 1991).The enzyme immunoassay can also be used <strong>to</strong> detect circulating antigens or <strong>to</strong>diagnose tuberculosis in homogenized animal tissues (Thoen et al., 1981). For a program<strong>to</strong> eliminate infected badgers, a serological procedure is being sought thatcould detect individual infected animals <strong>and</strong> thus prevent indiscriminate slaughter.In Australia, a simple test has been developed <strong>to</strong> measure in vitro the cell-mediatedimmune response <strong>to</strong> bovine PPD tuberculin. The test is based on detecting—using a s<strong>and</strong>wich enzyme immunoassay—gamma-interferon produced by incubation(for 24 hours) of whole bovine blood in the presence of tuberculin (Rothel etal., 1990). A field study conducted of a large number of cattle compared the analcaudaltest with PPD tuberculin <strong>and</strong> the gamma-interferon assay. Specificity withgamma-interferon was 96% <strong>to</strong> 98%, while sensitivity was 76.8% <strong>to</strong> 93.6% (dependingon the method of interpretation). If the two diagnostic tests are combined, it ispossible <strong>to</strong> obtain sensitivity of 95.2% (Wood et al., 1991; Wood et al., 1992). Thes<strong>and</strong>wich enzyme immunoassay <strong>to</strong> detect gamma-interferon in whole bovine bloodproved <strong>to</strong> be more sensitive <strong>and</strong> specific than the direct enzyme immunoassay fordetecting IgG in serum. In a study conducted in Argentina, the gamma-interferontest was positive in 9 of 19 animals that had tuberculous lesions limited <strong>to</strong> the lymphnodes <strong>and</strong> no antibodies in the ELISA test. In contrast, cattle with disseminatedlesions had a high antibody titer <strong>and</strong> little or no gamma-interferon production(Ritacco et al., 1991).Control: Prevention of human infection by M. bovis consists of the pasteurizationof milk, vaccination with BCG, <strong>and</strong> above all, control <strong>and</strong> eradication of bovinetuberculosis.The only rational approach <strong>to</strong> reducing <strong>and</strong> eliminating losses produced by theinfection in cattle <strong>and</strong> preventing human cases caused by M. bovis consists of establishinga control <strong>and</strong> eradication program for bovine tuberculosis. Eradication campaignsare usually carried out by administering tuberculin tests repeatedly, until allinfected animals are eliminated from the herd. Application of the tuberculin test <strong>and</strong>slaughter of reac<strong>to</strong>rs has given excellent results in all countries that have undertakeneradication campaigns. At present, many developed countries are free or practicallyfree of bovine tuberculosis. In developing countries, the inability of governments <strong>to</strong>


296 BACTERIOSEScompensate owners for the destruction of reac<strong>to</strong>rs hinders establishment of eradicationprograms <strong>and</strong> makes it necessary <strong>to</strong> find other incentives, such as a surchargeon milk. Campaigns should be begun in regions of low prevalence, where replacingreacting animals is easier, <strong>and</strong> later extended <strong>to</strong> areas of higher prevalence. The successof a program depends on the cooperation of the meat inspection agencies so thattuberculosis-free herds are correctly certified, activities are evaluated, <strong>and</strong> appropriateepidemiologic surveillance is maintained. The cooperation of the health servicesis also important <strong>to</strong> prevent persons with tuberculosis from working with animals<strong>and</strong> either infecting or sensitizing them.Controlling tuberculosis caused by M. bovis in its principal reservoir, cattle, is thebest method of preventing transmission <strong>to</strong> other species, including man.Several vaccines have been tested for preventing bovine tuberculosis, among themBCG, but none have proved effective. Treatment with anti-tuberculosis drugs, particularlyisoniazid, takes many months, is costly, can produce drug-resistant M.bovis strains, <strong>and</strong> the result is uncertain.Data on the status of bovine tuberculosis in Latin America <strong>and</strong> the Caribbean,with a summary on other countries, have been compiled <strong>and</strong> tabulated by de Kan<strong>to</strong>r<strong>and</strong> Alvarez (1991) <strong>and</strong> de Kan<strong>to</strong>r <strong>and</strong> Ritacco (1994).BibliographyArgentina, Comisión Nacional de Zoonosis, Subcomisión de Tuberculosis Bovina. Latuberculosis bovina en la República Argentina. Buenos Aires: Centro Panamericano deZoonosis; 1982.Berggren, S.A.. Field experiment with BCG vaccine in Malawi. Br Vet J 137:88–96, 1981.Cited in: Pritchard, D.G. A century of bovine tuberculosis 1888–1988: Conquest <strong>and</strong> controversy.J Comp Pathol 99:357–399, 1988.Burjanova, B., M. Nagyova. [Tuberculosis due <strong>to</strong> Mycobacterium bovis in the human populationin Slovakia 1978–1983]. Studia Pneumologica Phtiseologica Cechoslavaca45:342–346, 1985.Casal, M., M.J. Linares. Enzymatic profile of Mycobacterium tuberculosis. Eur J ClinMicrobiol 3:155–156, 1985.Castets, M., N. Rist, H. Boisvert. Le varieté africaine du bacille tuberculeux. Medicine del’Afrique Noire 16:321, 1969.Centrángolo, A., L.S. de Marchesini, C. Isola, I.N. de Kan<strong>to</strong>r, M. Di Lonardo. ElMycobacterium bovis como causa de tuberculosis humana. Actas, 13r. Congreso Argentino deTisiología, Mar del Plata, 1973.Cheeseman, C.L., J.W. Wilesmith, F.A. Stuart. Tuberculosis: The disease <strong>and</strong> its epidemiologyin the badger, a review. Epidemiol Infect 103:113–125, 1989.Collins, D.M., G.W. De Lisle. DNA restriction endonuclease analysis of Mycobacteriumbovis <strong>and</strong> other members of the tuberculosis complex. J Clin Microbiol 21:562–564, 1985.Collins, D.M., D.M. Gabric, G.W. De Lisle. Typing of Mycobacterium bovis isolates fromcattle <strong>and</strong> other animals in the same locality. N Z Vet J 36:45–46, 1988.Collins, C.H., J.M. Grange. The bovine tubercle bacillus. J Appl Bacteriol 55:13–29, 1983.Collins, C.H., M.D. Yates, J.M. Grange. A study of bovine strains of Mycobacterium tuberculosisisolated from humans in South-East Engl<strong>and</strong>, 1977–1979. Tubercle 62:113–116, 1981.Cordes, D.O., J.A. Bullians, D.E. Lake, M.E. Carter. Observations on tuberculosis causedby Mycobacterium bovis in sheep. N Z Vet J 29:60–62, 1981.Davidson, R.M., M.R. Alley, N.S. Beatson. Tuberculosis in a flock of sheep. N Z Vet J29:1–2, 1981.


ZOONOTIC TUBERCULOSIS 297de Kan<strong>to</strong>r, I.N., E. Alvarez, eds. Current status of bovine tuberculosis in Latin America <strong>and</strong>the Caribbean. Buenos Aires: Pan American Zoonoses Center; 1991. (Special Publication 10).de Kan<strong>to</strong>r, I.N., L. Barrera, V. Ritacco, I. Miceli. Utilidad del enzimoinmunoensayo en eldiagnóstico de la tuberculosis. Bol Oficina Sanit Panam 110:461–470, 1991.de Kan<strong>to</strong>r, I.N., E. de la Vega, A. Bernardelli. Infección por Mycobacterium bovis en liebresen la provincia de Buenos Aires, Argentina. Rev Med Vet 65:268–279, 1984.de Kan<strong>to</strong>r, I.N., N. Marchevsky, I.W. Lesslie. Respuesta al PPD en pacientes tuberculososafectados por M. tuberculosis y por M. bovis. Medicina 36:127–130, 1976.de Kan<strong>to</strong>r, I.N., J. Pereira, J. Miquet, R. Rovère. Pouvoir pathogène experimental deMycobacterium africanum pour les bovins. Bull Acad Vet Fr 52:499–503, 1979.de Kan<strong>to</strong>r, I.N., V. Ritacco. Bovine tuberculosis in Latin America <strong>and</strong> the Caribbean:Current status, control <strong>and</strong> eradication programs. Vet Microbiol 40:5–14, 1994.De Lisle, G.W., D.M. Collins, A.S. Loveday, et al. A report of tuberculosis in cats in NewZeal<strong>and</strong> <strong>and</strong> the examination of strains of Mycobacterium bovis by DNA restriction endonucleaseanalysis. N Z Vet J 38:10–13, 1990.Essey, M.A., A. Fanning, D.A. Saavi, J. Payeur. Bovine tuberculosis in Cervidae; humanhealth concerns. Proc Annu Meet U S Anim Health Assoc 95:427–436, 1991.Feldman, W.H. Tuberculosis. In: Hull, T.G., ed. Diseases Transmitted from Animals <strong>to</strong> Man.5th ed. Springfield: Thomas; 1963.Fernández Salazar, M., V. Gómez P<strong>and</strong>o, L. Domínguez Paredes. Mycobacterium bovis enla pa<strong>to</strong>logía humana en el Perú. Bol Inf Col Med Vet 14:16–18, 1983.Francis, J. Tuberculosis in Animals <strong>and</strong> Man. A Study in Comparative Pathology. London:Cassel; 1958.Francis, J., C.L. Choi, A.J. Frost. The diagnosis of tuberculosis in cattle with special reference<strong>to</strong> bovine PPD tuberculin. Aust Vet J 49:246–251, 1973.García Carrillo, C., B. Szyfres. La tuberculosis animal en las Américas y su transmisiónal hombre. Roma: Organización de las Naciones Unidas para la Agricultura y la Alimentación;1963.Good, R.C., D.E. Snider, Jr. Isolation of nontuberculous mycobacteria in the United States,1980. J Infect Dis 146:829–833, 1982.Grange, J.M., C.H. Collins. Bovine tubercle bacilli <strong>and</strong> disease in animals <strong>and</strong> man.Epidemiol Infect 99:221–234, 1987.Grange, J.M., M.D. Yates. Incidence <strong>and</strong> nature of human tuberculosis due <strong>to</strong>Mycobacterium africanum in South-East Engl<strong>and</strong>: 1977–87. Epidemiol Infect 103:127–132, 1989.Griffith, A.S. Bovine tuberculosis in man. Tubercle 18:528–543, 1937. Cited in: Collins,C.H., J.M. Grange. The bovine tubercle bacillus. J Appl Bacteriol 55:13–29, 1983.Hawthorne, V.M., I.M. Lauder. Tuberculosis in man, dog <strong>and</strong> cat. Am Rev Resp Dis85:858–869, 1962.Horsburgh, C.R., U.G. Mason III, D.C. Farhi, M.D. Iseman. Disseminated infection withMycobacterium avium-intracellulare. A report of 13 cases <strong>and</strong> review of the literature.Medicine 64:36–48, 1985.Huitema, H. Development of a comparative test with equal concentrations of avian <strong>and</strong>bovine PPD tuberculin for cattle. Tijdschr Diergeneeskd 98:396–407, 1973.Huitema, H. The eradication of bovine tuberculosis in cattle in the Netherl<strong>and</strong>s <strong>and</strong> the significanceof man as a source of infection in cattle. Selected Papers of the Royal Netherl<strong>and</strong>sTuberculosis Association 12:62–67, 1969. Cited in: Grange, J.M., C.H. Collins. Bovine tuberclebacilli <strong>and</strong> disease in animals <strong>and</strong> man. Epidemiol Infect 99:221–234, 1987.Humble, M.W., A. King, I. Phillips. API ZYM: A simple rapid system for the detection ofbacterial enzymes. J Clin Pathol 30:275–277, 1977.Isaac, J., J. Whitehead, J.W. Adams, M.D. Bar<strong>to</strong>n, P. Coloe. An outbreak of Mycobacteriumbovis infection in cats in an animal house. Aust Vet J 60:243–245, 1983.


298 BACTERIOSESJoint FAO/<strong>WHO</strong> Expert Committee on Zoonoses. Joint FAO/<strong>WHO</strong> Expert Committee onZoonoses. Third report. Geneva: <strong>WHO</strong>; 1967. (Technical Report Series 378).Karlson, A.G. Tuberculosis. In: Dunne, H.W., ed. Diseases of Swine. 3rd ed. Ames: IowaState University Press; 1970.Kleeberg, H.H. Tuberculosis <strong>and</strong> other mycobacterioses. In: Hubbert, W.T., W.F.McCulloch, P.R. Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed.Springfield: Thomas; 1975.Konyha, L.D., J.P. Kreier. The significance of tuberculin tests in the horse. Am Rev RespDis 103:91–99, 1971.Kubin, M., J. Stastna, M. Havelkova. [Classification of mycobacterial causal agents oftuberculosis <strong>and</strong> mycobacteriosis in the Czech Republic in 1981–1983]. Studia PneumologicaPhtisiologica Cechoslovaca 45:599–603, 1985.Lesslie, I.W., K.J. Birn, P. Stuart, et al. Tuberculosis in the pig <strong>and</strong> the tuberculin test. VetRec 83:647–651, 1968.Liu, S., I. Weitzman, G.G. Johnson. Canine tuberculosis. J Am Vet Med Assoc 177:164–167, 1980.Magnus, K. Epidemiological basis of tuberculosis eradication. 3. Risk of pulmonary tuberculosisafter human <strong>and</strong> bovine infection. Bull World Health Organ 35:483–508, 1966.Magnus, K. Epidemiological basis of tuberculosis eradication. 5. Frequency of pulmonarycalcifications after human <strong>and</strong> bovine infection. Bull World Health Organ 36:703–718, 1967.Matthias, D. Vergleichende Pathologie der Tuberkulose der Tiere. In: Meissner, G., A.Schmiedel, eds. Mykobakterien und Mykobakterielle Krankheiten. Teil VII. Jena: Fischer; 1970.McAleer, P.D. The relationship between badger density <strong>and</strong> the incidence of bovine tuberculosisin County Galway. Ir Vet J 43:77–80, 1990.Myers, J.A., J.H. Steele. Bovine Tuberculosis Control in Man <strong>and</strong> Animals. St. Louis:Green; 1969.Pan American Health Organization (<strong>PAHO</strong>). First International Seminar on BovineTuberculosis for the Americas, Santiago, Chile, 21–25 September 1970. Washing<strong>to</strong>n, D.C.:<strong>PAHO</strong>; 1972. (Scientific Publication 258).Pas<strong>to</strong>ret, P.P., E.T. Thiry, B. Brochier, et al. Maladies de faune sauvage transmissibles auxanimaux domestiques. Rev Sci Tech Off Int Epiz 7:661–704, 1988.Patterson, A.B., J.T. Stamp, J.N. Ritchie. Tuberculosis. In: Stableforth, A.W., I.A.Galloway, eds. Infectious Diseases of Animals. London: Butterworths; 1959.Pritchard, D.G. A century of bovine tuberculosis 1888–1988: Conquest <strong>and</strong> controversy. JComp Pathol 99:357–399, 1988.Ritacco, V., B. López, L. Barrera, et al. Further evaluation of an indirect enzyme-linkedimmunosorbent assay for the diagnosis of bovine tuberculosis. Zentralbl Veterinarmed [B]37:19–27, 1990.Ritacco, V., B. López, I.N. de Kan<strong>to</strong>r, et al. Reciprocal cellular <strong>and</strong> humoral immuneresponses in bovine tuberculosis. Res Vet Sci 50:365–367, 1991.Robinson, R.C., P.H. Phillips, G. Stevens, P.A. S<strong>to</strong>rm. An outbreak of Mycobacterium bovisin fallow deer (Dama dama). Aust Vet J 66:195–197, 1989.Roswurm, J.D., L.D. Konyha. The comparative-cervical tuberculin test as an aid <strong>to</strong> diagnosingbovine tuberculosis. Proc Annu Meet U S Anim Health Assoc 77:368–389, 1973.Rothel, J.S., S.L. Jones, L.A. Corner, et al. A s<strong>and</strong>wich enzyme immunoassay for bovineinterferon-gamma <strong>and</strong> its use for the detection of tuberculosis in cattle. Aust Vet J 67:134–137, 1990.Ruch, T.C. Diseases of Labora<strong>to</strong>ry Primates. Philadelphia: Saunders; 1959.Schliesser, T. Epidemiologie der Tuberkulose der Tiere. In: Meissner, G., A. Schmiedel,eds. Mykobakterien und Mykobakterielle Krankheiten. Teil VII. Jena: Fischer; 1970.Schmiedel, A. Erkrankungen der Menschen durch Mycobacterium bovis. In: Meissner,G., A. Schmiedel, eds. Mykobakterien und Mykobakterielle Krankheiten. Teil VII. Jena:Fischer, 1970.


ZOONOTIC TUBERCULOSIS 299Schonfeld, J.K. Human-<strong>to</strong>-human spread of infection by M. bovis. Tubercle 63:143–144, 1982.Shoemaker, S.A., J.H. Fisher, W.D. Jones, C.H. Scoggin. Restriction fragment analysis ofchromosomal DNA defines different strains of Mycobacterium tuberculosis. Am Rev RespirDis 134:210–213, 1986.Sjorgen, I., I. Sutherl<strong>and</strong>. Studies of tuberculosis in man in relation <strong>to</strong> infection in cattle.Tubercle 56:127–133, 1974.Stuart, F.A., P.A. Manser, F.G. McIn<strong>to</strong>sh. Tuberculosis in imported red deer (Cervus elaphus).Vet Rec 122:508–511, 1988.Thoen, C.O. Tuberculosis. In: Leman, A.D., B.E. Straw, W.L. Mengeling, S. D’Allaire, D.J.Taylor, eds. Diseases of Swine. 7th ed. Ames: Iowa State University; 1992.Thoen, C.O., C. Malstrom, E.M. Himes, K. Mills. Use of enzyme-linked immunosorbentassay for detecting mycobacterial antigens in tissues of Mycobacterium bovis-infected cattle.Am J Vet Res 42:1814–1815, 1981.Thorel, M.F. Isolation of Mycobacterium africanum from monkeys. Tubercle 61:101–104, 1980.Veen, J., J.V. Kuyvenhoven, E.T.B. Dinkla, et al. [Tuberculosis in alpacas; a zoonosis as animported disease]. Nederl<strong>and</strong>s Tijdschr Geneesk 135:1127–1130, 1991.Vestal, A.L. Procedures for the isolation of Mycobacteria. Atlanta: Centers for DiseaseControl <strong>and</strong> Prevention; 1969. (Public Health Publication 1995).Wiessmann, J. Die Rindertuberkulose beim Menschen und ihre epidemiologischeBedentung fur die Veterinarmedizin. Schweiz Arch Tierhenlkd 102:467–471, 1960.Wilesmith, J.W. Epidemiological features of bovine tuberculosis in cattle herds in GreatBritain. J Hyg 90:159–176, 1983.Wilesmith, J.W., P.E. Sayers, R. Bode, et al. Tuberculosis in East Sussex. II. Aspects ofbadger ecology <strong>and</strong> surveillance for tuberculosis in badger populations (1976–1984). J Hyg97:11–26, 1986.Winkler, W.G., N.B. Gale. Tuberculosis. In: Davis, J.W., L.H. Karstad, D.O. Trainer, eds.Infectious Diseases of Wild Mammals. Ames: Iowa State University Press; 1970.Wood, P.R., L.A. Corner, J.S. Rothel, et al. Field comparison of the interferon-gamma assay<strong>and</strong> the intradermal tuberculin tests for the diagnosis of bovine tuberculosis. Aust Vet J68:286–290, 1991.Wood, P.R., L.A. Corner, J.S. Rothel, et al. A field evaluation of serological <strong>and</strong> cellulardiagnostic tests for bovine tuberculosis. Vet Microbiol 31:71–79, 1992.Yates, M.D., J.M. Grange. Incidence <strong>and</strong> nature of human tuberculosis due <strong>to</strong> bovine tuberclebacilli in South-East Engl<strong>and</strong>: 1977–1987. Epidemiol Infect 101:225–229, 1988.


Part IIMYCOSES


ADIASPIROMYCOSISICD-10 B48.8Synonyms: Adiaspirosis, haplomycosis, haplosporangiosis.Etiology: Chrysosporium (Emmonsia) parvum var. crescens <strong>and</strong> C. parvum var.parvum, saprophytic soil fungi that characteristically form large spherules (adiaspores)in the lungs. In the tissular phase, the fungus does not multiply. C. crescens isthe most <strong>common</strong> agent in man <strong>and</strong> animals. C. parva occurs primarily in animals <strong>and</strong>forms smaller spherules than C. crescens. C. parvum var. crescens <strong>and</strong> C. parvum var.parvum differ in size. In the lungs, C. crescens measures 200 <strong>to</strong> 700 microns, whereasC. parvum measures 40 microns. In addition, C. parvum is mononuclear, even whenit reaches its maximum size, while C. crescens eventually has hundreds of nuclei.Geographic Distribution: Worldwide. In the Americas, the infection has beenconfirmed in Argentina, Brazil, Canada, Guatemala, Honduras, the United States,<strong>and</strong> Venezuela.Occurrence in Man: Rare. Eleven human cases have been reported in Asia,Europe, South America, <strong>and</strong> the United States (Englund <strong>and</strong> Hochholzer, 1993).According <strong>to</strong> Moraes et al. (1989) there were 23 cases, four of which were extrapulmonary.Occurrence in Animals: Frequent in small wild mammals. The disease has beenconfirmed in at least 124 mammalian species or subspecies (Leigh<strong>to</strong>n <strong>and</strong> Wobeser,1978). Among other mammals, the disease has been diagnosed in skunks (Mephitismephitis) in Argentina, Canada, <strong>and</strong> the United States.The Disease in Man <strong>and</strong> Animals: The only clinically significant form, in manas well as animals, is pulmonary adiaspiromycosis. The few human cases have beendiagnosed through biopsy or au<strong>to</strong>psy specimens. The fungus causes light gray <strong>to</strong>yellowish lesions in the lungs, without greatly affecting the animal’s overall health.The number of spherules (adiaspores) in the lung tissue depends on the number ofconidia (spores) inhaled. In the lungs, the fungus increases significantly in size. Iffew conidia are inhaled, usually only one lung is affected. If the inoculum is large,both lungs are likely <strong>to</strong> be affected. Adiaspiromycosis usually disappears spontaneouslybut requires surgical resection if it persists (Englund <strong>and</strong> Hochholzer, 1993).The etiologic agent may also be found in other organs, though rarely. One case ofdisseminated adiaspiromycosis was described in an AIDS patient. The most significantclinical characteristic was disseminated osteomyelitis. The fungus,Chrysosporium parvum var. parvum, was isolated during surgery from the pus of awrist lesion, as well as from the sputum <strong>and</strong> bone-marrow aspirate. The mycoticinfection was controlled with amphotericin B (Echeverría et al., 1993). A fatal caseof adiaspiromycosis was recorded in Brazil in a 35-year-old rural worker who hadcomplained of generalized weakness, dry cough, afternoon fever, <strong>and</strong> a weight lossof 8 kg during the four weeks prior <strong>to</strong> hospitalization. Clinical symp<strong>to</strong>ms <strong>and</strong> radiographywere similar <strong>to</strong> miliary tuberculosis. The fungus was detected in the specimensobtained during au<strong>to</strong>psy (Peres et al., 1992). Another similar fatal case hadoccurred previously in Brazil (Moraes et al., 1989).303


304 MYCOSESThe disease is generally asymp<strong>to</strong>matic, but resection of the affected tissue may benecessary if it persists <strong>and</strong> symp<strong>to</strong>ms appear.In 7 out of 25 skunks (Mephitis mephitis) captured <strong>and</strong> au<strong>to</strong>psied in Alberta,Canada, lesions were found that varied from slight <strong>and</strong> only visible microscopically<strong>to</strong> severe with grayish-white nodules in the pulmonary parenchyma that spread <strong>to</strong>the bronchotracheal <strong>and</strong> mediastinal lymphatic ganglia. His<strong>to</strong>logically, the lesionswere characterized by a centrally located spherule surrounded by granuloma<strong>to</strong>usinflammation (Albassam et al., 1986).Source of Infection <strong>and</strong> Mode of Transmission: The great preponderance ofpulmonary localizations indicates that the infection is contracted through inhalation.C. crescens has been isolated from the soil. Differences in the infection ratesfor three very similar species of squirrels indicate that the fungus may be presentin certain habitats (Leigh<strong>to</strong>n <strong>and</strong> Wobeser, 1978), possibly linked <strong>to</strong> the rootmicroflora of certain plants. Other authors (cited by Mason <strong>and</strong> Gauhwin, 1982)suggest that preda<strong>to</strong>r-prey interactions affect its distribution: upon ingestinginfected animals, carnivores eliminate adiaspores in their feces, where the sporesgerminate <strong>and</strong> develop. This was demonstrated in cats, in a mustelid (Mustelanivalis), <strong>and</strong> in birds of prey. Thus, preda<strong>to</strong>rs could play a role in disseminating theetiologic agent.Under very windy conditions, both animals <strong>and</strong> humans may inhale airborne conidiareleased from the soil.Role of Animals: The soil is the reservoir for the fungus <strong>and</strong> the source of infectionin humans <strong>and</strong> other animals. It is believed that some animals may play a rolein disseminating the agent.Diagnosis: Diagnosis may be made by observation of spherules in lung tissue, bystained his<strong>to</strong>logical preparations, <strong>and</strong> by culture <strong>and</strong> inoculation in<strong>to</strong> labora<strong>to</strong>ry animals.The most effective method for detecting adiaspores in the lungs of animals istissue digestion with a 2% sodium hydroxide solution (Leigh<strong>to</strong>n <strong>and</strong> Wobeser,1978). The spherules are stained with acid-Schiff <strong>and</strong> Gomori methenamine silvernitrate reagents (Englund <strong>and</strong> Hochholzer, 1993).BibliographyAinsworth, G.C., P.K.C. Austwick. Fungal Diseases of Animals. 2nd ed. Farnham Royal,Slough, United Kingdom: Commonwealth Agricultural Bureau; 1973.Albassam, M.A., R. Bhatnagar, L.E. Lillie, L. Roy. Adiaspiromycosis in striped skunks inAlberta, Canada. J Wildl Dis 22:13–18, 1986.Cueva, J.A., M.D. Little. Emmonsia crescens infection (adiaspiromycosis) in man inHonduras. Am J Trop Med Hyg 20:282–287, 1971.Echevarría, E., E.L. Cano, A. Restrepo. Disseminated adiaspiromycosis in a patient withAIDS. J Med Vet Mycol 31:91–97, 1993.Engl<strong>and</strong>, D.M., L. Hochholzer. Adiaspiromycosis: An unusual fungal infection of the lung.Report of 11 cases. Am J Surg Pathol 17:876–886, 1993.Jellison, W.L. Adiaspiromycosis. In: Davis, J.W., L.H. Karstad, D.O. Trainer, eds.Infectious Diseases of Wild Mammals. Ames: Iowa State University Press; 1970.Leigh<strong>to</strong>n, F.A., G. Wobeser. The prevalence of adiaspiromycosis in three sympatric speciesof ground squirrels. J Wildl Dis 14:362–365, 1978.


ASPERGILLOSIS 305Mason, R.W., M. Gauhwin. Adiaspiromycosis in South Australian hairy-nosed wombats(Lasiorhinus latifrons). J Wildl Dis 18:3–8, 1982.Moraes, N.A., M.C. de Almeida, A.N. Raick. Caso fatal de adiaspiromicose pulmonarhumana. Rev Inst Med Trop Sao Paulo 31:188–194, 1989.Peres, L.C., F. Figueiredo, M. Peinado, F.A. Soares. Fulminant disseminated pulmonaryadiaspiromycosis in humans. Am J Trop Med Hyg 46:146–150, 1992.Salfelder, K. New <strong>and</strong> un<strong>common</strong> opportunistic fungal infections. In:Pan American HealthOrganization. Proceedings of the Third International Conference on the Mycoses.Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1975. (Scientific Publication 304).ASPERGILLOSISICD-10 B44.0 invasive pulmonary aspergillosis;B44.1 other pulmonary aspergillosis;B44.7 disseminated aspergillosis; B44.8 other forms of aspergillosisSynonyms: Pneumonomycosis, bronchomycosis (in animals).Etiology: Aspergillus fumigatus <strong>and</strong> occasionally other species of the genusAspergillus, such as A. flavus, A. nidulans, A. niger, <strong>and</strong> A. terreus. These saprophyticfungi are <strong>common</strong> components of the soil microflora; they play an importantrole in the decomposition of organic matter.Aspergillus flavus <strong>and</strong> A. parasiticus are known for their production of afla<strong>to</strong>xinsin oleaginous grains <strong>and</strong> seeds such as corn, rice, peanuts, <strong>and</strong> cot<strong>to</strong>nseeds s<strong>to</strong>redunder damp conditions. Afla<strong>to</strong>xin B 1is hepa<strong>to</strong><strong>to</strong>xic <strong>and</strong> carcinogenic for humans<strong>and</strong> animals. These fungi do not produce the afla<strong>to</strong>xin in animal tissue. Thus, thischapter covers only infection by Aspergillus spp.Geographic Distribution: The fungus is ubiqui<strong>to</strong>us <strong>and</strong> distributed worldwide.The disease has no particular distribution.Occurrence in Man: Aspergillosis occurs sporadically <strong>and</strong> is un<strong>common</strong>. Itsincidence, as is that of other opportunistic mycoses 1 (c<strong>and</strong>idiasis, zygomycosis), isincreasing due <strong>to</strong> the growing use of antibiotics, antimetabolites, <strong>and</strong> corticosteroids.It occurs frequently in advanced cases of cancer. Small nosocomial outbreakshave also been reported (see section on the disease in man).In Mexico, aspergillosis lesions were found in 1.2% of more than 2,000 r<strong>and</strong>omau<strong>to</strong>psies performed in a general hospital (González-Mendoza, 1970).Occurrence in Animals: Sporadic cases have been described in many species ofdomestic <strong>and</strong> wild mammals <strong>and</strong> birds. The disease in fowl <strong>and</strong> cattle has economic1Mycoses that attack debilitated persons or those treated over a long period with antibiotics,antimetabolites, or corticosteroids.


306 MYCOSESimplications. The incidence is low in adult domestic fowl, but outbreaks in chicks<strong>and</strong> young turkeys can cause considerable losses on some farms.The Disease in Man: Aspergillosis establishes itself in patients debilitated bychronic <strong>diseases</strong> (such as diabetes, cancer, tuberculosis, deep mycoses) <strong>and</strong> <strong>diseases</strong>of the immune system, as well as in persons treated with antibiotics, antimetabolites,<strong>and</strong> corticosteroids for prolonged periods. Persons occupationally exposed for longperiods <strong>to</strong> materials contaminated by fungus spores (grain, hay, cot<strong>to</strong>n, wool, <strong>and</strong>others) run a greater risk.A study group on aspergillosis in AIDS patients conducted a retrospective reviewof 33 patients with invasive aspergillosis in different medical facilities in France.Of this group of 33 patients, 91% were recorded from 1989 <strong>to</strong> 1991, suggestingthat invasive aspergillosis is an emerging complication of AIDS. Aspergillus spp.cultures were obtained from bronchopulmonary lavage of 28 patients, <strong>and</strong> no otherpathogenic agents were found. Of 15 patients who underwent biopsy or au<strong>to</strong>psy,14 were his<strong>to</strong>logically positive. The clinical <strong>and</strong> radiological symp<strong>to</strong>ms werecomparable <strong>to</strong> aspergillosis in non-AIDS patients with neutropenia, though theAIDS patients had a higher incidence of neurological complications (Lortholaryet al., 1993).There are two differentiated clinical forms of the disease: localized <strong>and</strong> invasive.Aspergillosis is essentially a respira<strong>to</strong>ry system infection acquired through inhalationof Aspergillus spp. conidia. Patients with pronounced granulocy<strong>to</strong>penia maycontract an acute <strong>and</strong> rapidly progressing pneumonia. The symp<strong>to</strong>ms are high fever,pulmonary consolidation, <strong>and</strong> cavitation. Normal children who inhale a large numberof conidia may develop fever, dyspnea, <strong>and</strong> miliary infiltration (Bennett, 1990).Allergic bronchopulmonary aspergillosis (ABPA) occurs in patients with preexistingasthma who present eosinophilia <strong>and</strong> intermittent bronchial obstruction(Bennett, 1990). Eosinophilia, precipitant antibodies, <strong>and</strong> high serum IgE concentrationare found in these patients; the intradermal [skin prick] test produces animmediate reaction <strong>to</strong> Aspergillus antigens, with papules <strong>and</strong> reddening. Despiterecurring exacerbations, some patients do not experience any permanent loss of pulmonaryfunction. Other patients, however, suffer corticoid-dependent asthma or permanen<strong>to</strong>bstructive disease (Bennett, 1990). ABPA patients may expec<strong>to</strong>ratebronchial plugs in which hyphae of the fungus can be detected microscopically.Even during remission, 33% of patients evidenced circulating immune complexes,primarily involving IgG (Bhatnagar et al., 1993).Allergic bronchopulmonary aspergillosis is more <strong>common</strong> than was thought in thepast. The disease may begin during childhood <strong>and</strong> continue without being clinicallyrecognized for many years or decades, until the patient begins <strong>to</strong> suffer from fibroticpulmonary disease. In this regard, it must be noted that aspergillosis infection maybe asymp<strong>to</strong>matic <strong>and</strong> suspected only due <strong>to</strong> a significant increase in serum IgE.Often the diagnosis comes <strong>to</strong>o late for chemotherapy treatment <strong>to</strong> be effective. Whencorticoids are discontinued, dyspnea <strong>and</strong> wheezing occur, requiring a return <strong>to</strong> medicationwith prednisolone (Greenberger, 1986). A later study concluded that inhaledbeclomethasone dipropionate may be more effective in treating ABPA than traditionalprednisolone by mouth (Imbeault <strong>and</strong> Cormier, 1993).Another form of the disease is the fungus ball or aspergilloma, which occurs whenthe fungus colonizes respira<strong>to</strong>ry cavities caused by other preexisting <strong>diseases</strong> (bron-


ASPERGILLOSIS 307chitis, bronchiectasis, tuberculosis). This form is relatively benign, but it occasionallyproduces hemoptysis.Other clinical forms are o<strong>to</strong>mycosis (often caused by A. niger) <strong>and</strong> invasion of theparanasal sinuses by the fungus. Though rare, the cutaneous form of the disease mayappear in immunodeficient patients.The invasive form is usually very serious. The fungi penetrate the blood vessels<strong>and</strong> can spread throughout the body. Cases of pulmonary aspergillosis have alsobeen described in patients who are not immunodeficient. There is general insistencethat the invasive form of the disease occurs only in patients with neutropenia.Neutrophil polymorphonuclear leukocytes are very important in the defense againstaspergillosis or in those who have serious defects in cell-mediated immunity (Karam<strong>and</strong> Griffin, 1986). Karam <strong>and</strong> Griffin describe three cases over five years in a universityhospital <strong>and</strong> cite 32 cases found in the literature. Of the 32 cases cited, 14had no underlying disease.Surgical intervention in the case of pulmonary or pleuropulmonary aspergillosismay be indicated <strong>to</strong> treat pleural empyemas <strong>and</strong> bronchopleural fistulae. In thesecases, myoplasty, thoracomyoplasty, <strong>and</strong> omen<strong>to</strong>plasty are the procedures most recommended(Wex et al., 1993). Surgical removal is also justified in the case of invasiveaspergillosis in the brain <strong>and</strong> paranasal sinuses, as well as in noninvasive colonizationof the paranasal sinuses (Bennett, 1990). When colonization is invasive, itis advisable <strong>to</strong> discontinue or reduce the use of immunosuppressants <strong>and</strong> <strong>to</strong> starttreatment with intravenous amphotericin B or itraconazole.Several small outbreaks have occurred during the renovation, expansion, orremodeling of hospitals <strong>and</strong> the construction of highways near hospitals. Duringthese projects, large numbers of conidia are made airborne, <strong>and</strong> may become concentrateddue <strong>to</strong> ventilation systems with defective filters. Between July 1981 <strong>and</strong>July 1988, 11 immunodeficient patients in a military hospital contracted disseminatedaspergillosis <strong>and</strong> died as a result. The hospital’s project involved the renovationof the intensive care unit <strong>and</strong> several other rooms. The infection spread no furtherafter several simultaneous measures were taken, such as installingfloor-<strong>to</strong>-ceiling partitions in the construction area, negative pressure in the samearea, antifungal decontamination using copper 8-hydroxyquinoline, <strong>and</strong> high-efficiencyparticulate air (HEPA) filters in air conditioning units <strong>and</strong> in rooms withimmunodeficient patients (Opal et al., 1986). However, a certain percentage ofpatients with lymphoma who received bone marrow transplants <strong>and</strong> were located insingle-occupancy rooms with positive air pressure <strong>and</strong> high efficiency air filters didacquire aspergillosis. Of 417 lymphoma patients studied, 22 (5.2%) contracted invasiveaspergillosis. These 22 patients were treated with amphotericin B, 17 of themprior <strong>to</strong> being diagnosed with aspergillosis; seven survived. All of the patients withdisseminated aspergillosis died (Iwen et al., 1993).The Disease in Animals: Although aspergillosis occurs sporadically in manyanimal species, where it primarily causes respira<strong>to</strong>ry system disorders, the followingdiscussion only deals with the disease in cattle, horses, dogs, <strong>and</strong> fowl.CATTLE: It is estimated that 75% of mycotic abortions are due <strong>to</strong> Aspergillus, particularlyA. fumigatus, <strong>and</strong> 10% <strong>to</strong> 15% <strong>to</strong> fungi of the order Mucorales. As brucellosis,campylobacteriosis, <strong>and</strong> trichomoniasis are brought under control, the relativerole of fungi as a cause of abortions increases. Mycotic abortion is seen mainly in


308 MYCOSESstabled animals; thus, it occurs during the winter in countries with cold or temperateclimates. Generally, only one or two females in a herd abort.The pathogenesis of the disease is not well known. It is thought that the fungusfirst localizes in the lungs or the digestive system, where it multiplies before invadingthe placenta via the bloodstream <strong>and</strong> causing placentitis. Most abortions occurduring the third trimester of pregnancy. The cotyledons swell <strong>and</strong> turn a brownishgray color. In serious cases, the placenta becomes wrinkled <strong>and</strong> leathery. The fungusmay invade the fetus as well, causing dermatitis <strong>and</strong> bronchopneumonia.Retention of the placenta is <strong>common</strong>. Other forms of the infection are the pulmonaryforms, also due primarily <strong>to</strong> A. fumigatus, <strong>and</strong> skin aspergillomas, caused by A. terreus(Schmitt, 1981).HORSES: Invasive pulmonary aspergillosis is relatively rare in horses. As in cattle,the disease is generally associated with abortion. There is also an associationbetween enterocolitis (Salmonella, Ehrlichia ristici) <strong>and</strong> invasive pulmonaryaspergillosis (Hattel et al., 1991).DOGS: Aspergillosis in dogs is generally confined <strong>to</strong> the nasal cavity or paranasalsinuses. A. fumigatus is the most <strong>common</strong> fungus. Disseminated aspergillosis is rare<strong>and</strong> has been found in dry, warm regions. In Australia, 12 cases due <strong>to</strong> A. terreuswere recorded during 1980–1984. Eleven of the 12 dogs were German Shepherds.The disease was characterized by granulomas in several organs, particularly in thekidneys, spleen, <strong>and</strong> bones. Lumbar diskospondylitis <strong>and</strong> focal osteomyelitis were<strong>common</strong>, generally in the epiphysis of the long bones (Day et al., 1986). Six casesof disseminated aspergillosis were recorded in the United States with characteristicssimilar <strong>to</strong> those in Australia (Dallman et al., 1992).FOWL: Outbreaks of acute aspergillosis occur in chicks <strong>and</strong> young turkeys, sometimescausing considerable losses. The symp<strong>to</strong>ms include fever, loss of appetite,labored breathing, diarrhea, <strong>and</strong> emaciation. In chronic aspergillosis, which occurssporadically in adult birds, the clinical picture is varied <strong>and</strong> depends on the localization.The affected birds may survive for a long time in a state of general debilitation.Yellowish granulomas of 1 <strong>to</strong> 3 mm (or larger, if the process is chronic) appearin the lungs. Plaques develop in the air sacks <strong>and</strong> may gradually cover the serosa;the same lesions or a mucoid exodate are found in the bronchial tubes <strong>and</strong> the trachea.Granuloma<strong>to</strong>us lesions are also found frequently in different organs, as eithernodules or plaques. The principal etiologic agents are A. fumigatus <strong>and</strong> A. flavus.Many species of domesticated <strong>and</strong> wild birds are susceptible <strong>to</strong> the disease. Captivepenguins frequently are victims (Chute <strong>and</strong> Richard, 1991).Clinical forms other than the pulmonary form occur in birds. These are dermatitis,osteomycosis, ophthalmitis, <strong>and</strong> encephalitis. Osteomycosis <strong>and</strong> encephalitis areprobably spread through the bloodstream (Chute <strong>and</strong> Richard, 1991).Source of Infection <strong>and</strong> Mode of Transmission: The reservoir is the soil. Theinfecting element is the conidia (exospores) of the fungus, which are transmitted <strong>to</strong>man <strong>and</strong> animals through the air. The causal agent is ubiqui<strong>to</strong>us <strong>and</strong> can survive inthe most varied environmental conditions. Despite this, the disease does not occurfrequently in man, indicating natural resistance <strong>to</strong> the infection. This resistance maybe undone by the use of immunosuppressant medications or fac<strong>to</strong>rs that impair theimmune system (see the section on the disease in man for other details on predis-


ASPERGILLOSIS 309posing fac<strong>to</strong>rs <strong>and</strong> <strong>diseases</strong>). In domestic mammals <strong>and</strong> birds, as well as in peoplewho work with them, an important source of infection is fodder <strong>and</strong> bedding contaminatedby the fungus, which releases conidia upon maturing. Apparently, exposuremust be prolonged or massive for the infection <strong>to</strong> become established. Airborneconidia are found in incuba<strong>to</strong>rs, hatcheries, incubation rooms, <strong>and</strong> air ducts; thesemay be the source of infection for chicks or young turkeys (Chute <strong>and</strong> Richard,1991).Role of Animals in Epidemiology: The source of infection is always the environment.The infection is not transmitted from one individual <strong>to</strong> another (man oranimal).Diagnosis: Due <strong>to</strong> the ubiqui<strong>to</strong>us nature of the agent, isolation by culture is not areliable test, since the agent may exist as a contaminant in the environment (labora<strong>to</strong>ryor hospital) or as a saprophyte in the upper respira<strong>to</strong>ry tract. A conclusive testmay be obtained by simultaneously conducting a his<strong>to</strong>logical examination usingbiopsy material <strong>and</strong> confirming the presence of the fungus in the preparations. Theagent may also be isolated by culturing aseptically obtained specimens from lesionsnot exposed <strong>to</strong> the environment. The species can only be identified by means of aculture. The immunodiffusion test has yielded very good results, as have counterimmunoelectrophoresis<strong>and</strong> enzyme-linked immunosorbent assay (ELISA).Serological tests are useful for diagnosing aspergillomas <strong>and</strong> in allergic bronchopulmonaryaspergillosis, but not in invasive aspergillosis (Bennett, 1990). Highlevels of A. fumigatus-specific IgE <strong>and</strong> IgG are detected in the sera of ABPApatients, while IgG alone is detected in aspergillomas (Kurup, 1986). In fowl, it isenough <strong>to</strong> confirm the presence of the fungus through direct observation or by culturingmaterials from lesions of sacrificed birds.Control: Due <strong>to</strong> the ubiqui<strong>to</strong>us nature of the fungus, it is impossible <strong>to</strong> establishpractical control measures. Prolonged treatment with antibiotics or corticosteroidsshould be limited <strong>to</strong> cases in which such therapy is essential. It is advisable <strong>to</strong> takespecial precautionary measures <strong>to</strong> avoid nosocomial outbreaks <strong>and</strong> <strong>to</strong> protectimmunodeficient patients when construction work is done inside or near hospitals.Patients with lymphoma who receive bone marrow transplants should receive prophylactictreatment with amphotericin B (Iwen et al., 1993). Moldy bedding or foddershould not be h<strong>and</strong>led or given <strong>to</strong> domestic mammals <strong>and</strong> birds. Hygienic conditionsin incuba<strong>to</strong>rs <strong>and</strong> incubation rooms are important in preventing avianaspergillosis.BibliographyAinsworth, G.C., P.K.C. Austwick. Fungal Diseases of Animals. 2nd ed. Farnham Royal,Slough, United Kingdom: Commonwealth Agricultural Bureau; 1973.Ajello, L., L.K. Georg, W. Kaplan, L. Kaufman. Labora<strong>to</strong>ry Manual for MedicalMycology. Washing<strong>to</strong>n, D.C.: U.S. Government Printing Office; 1963. (Public Health ServicePublication 994).Bennett, J.E. Aspergillus species. In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E. Bennett, eds.Vol 2: Principles <strong>and</strong> Practice of Infectious Disease. 3rd ed. New York: Churchill Livings<strong>to</strong>ne,Inc.; 1990.Bhatnagar, P.K., B. Banerjee, P.V. Sarma. Serological findings in patients with allergicbronchopulmonary aspergillosis during remission. J Infect 27:33–37, 1993.


310 MYCOSESChute, H.L., J.L. Richard. Fungal infections. In: Calnek, B.W., H.J. Barnes, C.W. Beard,W.M. Reid, H.W. Yoder, Jr., eds. Diseases of Poultry. 9th ed. Ames: Iowa State UniversityPress; 1991.Dallman, M.J., T.L. Dew, L. Tobias, R. Doss. Disseminated aspergillosis in a dog withdiskospondylitis <strong>and</strong> neurologic deficits. J Am Vet Med Assoc 200:511–513, 1992.Day, M.J., W.J. Penhale, C.E. Eger, et al. Disseminated aspergillosis in dogs. Aust Vet J63:55–59, 1986.González-Mendoza, A. Opportunistic mycoses. In: Pan American Health Organization.Proceedings: International Symposium on Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1970.(Scientific Publication 205).Gordon, M.A. Current status of serology for diagnosis <strong>and</strong> prognostic evaluation of opportunisticfungus infections. In: Pan American Health Organization. Proceedings of the ThirdInternational Conference on the Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1975. (ScientificPublication 304).Greenberger, P.A. Aspergillosis: Clinical aspects. Zbl Bakt Hyg A 261:487–495, 1986.Hattel, A.L., T.R. Drake, B.J. Anderholm, E.S. McAllister. Pulmonary aspergillosis associatedwith acute enteritis in a horse. J Am Vet Med Assoc 199:589–590, 1991.Imbeault, B., Y. Cormier. Usefulness of inhaled high-dose corticosteroids in allergic bronchopulmonaryaspergillosis. Chest 103:1614–1617, 1993.Iwen, P.C., E.C. Reed, J.O. Armitage, et al. Nosocomial invasive aspergillosis in lymphomapatients treated with bone marrow or peripheral stem cell transplants. Infect Control HospEpidemiol 14:131–139, 1993.Karam, G.H., F.M. Griffin, Jr. Invasive pulmonary aspergillosis in nonimmunocompromised,nonneutropenic hosts. Rev Infect Dis 8:357–363, 1986.Kurup, V.P. Enzyme-linked immunosorbent assay in the detection of specific antibodiesagainst Aspergillus in patient sera. Zbl Bakt Hyg A 261:509–516, 1986.Lortholary, O., M.C. Meyohas, B. Dupont, et al. Invasive aspergillosis in patients withacquired immunodeficiency syndrome: Report of 33 cases. French Cooperative Study Groupon Aspergillosis in AIDS. Am J Med 95:177–187, 1993.Mishra, S.K., S. Falkenberg, N. Masihi. Efficacy of enzyme-linked immunosorbent assayin serodiagnosis of aspergillosis. J Clin Microbiol 17:708–710, 1983.Opal, S.M., A.A. Asp, P.B. Cannady, Jr., et al. Efficacy of infection control measures duringa nosocomial outbreak of disseminated aspergillosis associated with hospital construction.J Infect Dis 153:634–637, 1986.Schmitt, J.A. Mycotic <strong>diseases</strong>. In: Ristic M., I. McIntyre, eds. Diseases of Cattle in theTropics. The Hague: Martinus Nijhoff; 1981.Utz, J.P. The systemic mycoses. In:Wyngaarden, J.B., L.H. Smith, Jr., eds. Cecil Textbookof Medicine. 16th ed. Philadelphia: Saunders; 1982.Wex, P., E. Utta, W. Drozdz. Surgical treatment of pulmonary <strong>and</strong> pleuropulmonaryAspergillus disease. Thorac Cardiovasc Surg 41:64–70, 1993.Winter, A.J. Mycotic abortion. In: Faulkner, L.C., ed. Abortion Diseases of Lives<strong>to</strong>ck.Springfield: Thomas; 1968.


BLASTOMYCOSIS 311BLASTOMYCOSISICD-10 B40.0 acute pulmonary blas<strong>to</strong>mycosis; B40.1 chronic pulmonaryblas<strong>to</strong>mycosis; B40.3 cutaneous blas<strong>to</strong>mycosis; B40.7 disseminatedblas<strong>to</strong>mycosis; B40.8 other forms of blas<strong>to</strong>mycosisSynonyms: North American blas<strong>to</strong>mycosis, Chicago disease, Gilchrist’s disease.Etiology: Blas<strong>to</strong>myces dermatitidis,a dimorphic fungus existing in mycelial formin cultures <strong>and</strong> as a budding yeast in the tissues of infected mammals. The fungusalso exists as yeast in enriched culture media at 37°C. The mycelial form in culturemedia at 25°C is cot<strong>to</strong>ny white, turning <strong>to</strong> brown over time.S<strong>and</strong>y, acidic soil close <strong>to</strong> rivers or other freshwater reservoirs is the microecosystemmost favorable <strong>to</strong> B. dermatitidis. It remains in an infective sporulatedstate in this bio<strong>to</strong>pe, as its spores (conidia) can detach <strong>and</strong> become airborne. Highambient humidity seems <strong>to</strong> favor the release of spores.B. dermatitidis is subdivided in<strong>to</strong> two serotypes (1 <strong>and</strong> 2) based on the presenceof an exoantigen, called A <strong>and</strong> recognized by a specific precipitin. Strains examinedfrom India, Israel, <strong>and</strong> the United States, <strong>and</strong> one strain examined from Africa allcontained A antigen (serotype 1). Eleven of 12 African strains examined were type2. The African strains are deficient in A antigen, but contain K antigen (Kaufman etal., 1983).Geographic Distribution: The disease has been observed in eastern Canada,India, Israel, South Africa, Tanzania, Tunisia, Ug<strong>and</strong>a, the United States, <strong>and</strong>the former Zaire. Au<strong>to</strong>chthonous cases have also occurred in some Central <strong>and</strong>South American countries (Klein et al., 1986). In the United States, endemic areasare located along the Mississippi, Missouri, <strong>and</strong> Ohio rivers, <strong>and</strong> in parts of NewYork State. In Canada, they are located along the St. Lawrence River <strong>and</strong> near theGreat Lakes.Occurrence in Man: Predominantly sporadic. Most of the cases have beenrecorded in the United States, with the highest prevalence in the Mississippi <strong>and</strong>Ohio river basins. From 1885 <strong>to</strong> 1968, there were 1,573 cases in that country(Menges as cited by Selby, 1975). Klein et al. (1986) summarized from the literaturethe incidence in different endemic U.S. states: from 0.1 <strong>to</strong> 0.7 cases per 100,000inhabitants per year in Arkansas from 1960 <strong>to</strong> 1965; 0.61, 0.44, <strong>and</strong> 0.43 cases per100,000 inhabitants per year in Mississippi, Kentucky, <strong>and</strong> Arkansas, respectively,from 1960 <strong>to</strong> 1967; <strong>and</strong> 0.48 cases per 100,000 inhabitants per year in Wisconsinfrom 1873 <strong>to</strong> 1982. Hyperendemic areas in these states have an incidence of 4 casesper 100,000 per year. These data do not include slight cases of the disease that donot generally receive medical attention.In Louisiana (USA), an attempt was made <strong>to</strong> identify all cases that occur in thestate <strong>and</strong> <strong>to</strong> study one district in detail (Washing<strong>to</strong>n Parish) that is consideredendemic. The average annual incidence for the entire state during 1976–1985 was0.23 cases per 100,000 inhabitants, while the incidence for Washing<strong>to</strong>n Parish was6.8 cases per 100,000. In 30 cases studied in this district, the patients’ ages rangedfrom 3 weeks <strong>to</strong> 81 years. Five people died, <strong>and</strong> one of these was probably infectedin utero (Lowry et al., 1989).


312 MYCOSESIn Canada, about 120 cases of blas<strong>to</strong>mycosis were recorded up until 1979. Mos<strong>to</strong>f the cases occurred in Quebec, followed by Ontario <strong>and</strong> the maritime provinces.More recently, 38 cases were reported in Ontario, as was a new focus <strong>to</strong> thenorth <strong>and</strong> east of Lake Superior that accounted for 20 of the patients (Bakerspigelet al., 1986).The disease also occurs in the form of outbreaks. Klein et al. (1986) reportedseven of them, mainly in the northern part of the mid-western United States. Thelargest outbreak affected 48 people in Wisconsin who traveled <strong>to</strong> a beaver pond <strong>and</strong>also visited their dens <strong>and</strong> dams. Only one outbreak occurred in an urban area (nearChicago), <strong>and</strong> in nine months affected five people living close <strong>to</strong> a highway underconstruction. Another outbreak in a wooded, marshy area of Virginia simultaneouslyaffected four raccoon hunters <strong>and</strong> their four hunting dogs (Armstrong et al., 1987).The disease occurs more frequently among males <strong>and</strong> the highest rate of infection isfound in men over 20 years of age. Most cases occur in winter (Klein et al., 1986).Occurrence in Animals: Sporadic. Canids are the most affected, <strong>and</strong> the greatestconcentration of cases is seen in Arkansas (USA). A study was conducted on theaccumulated data of 20 university veterinary hospitals in terms of the risk fac<strong>to</strong>rs forblas<strong>to</strong>mycosis in dogs. From 1980 <strong>to</strong> 1990, 971 cases were recorded. The prevalenceof blas<strong>to</strong>mycosis in dogs was 205 per 100,000 hospital admissions. The highest incidenceof the disease occurred in autumn. The principal victims were hunting dogsweighing between 23 <strong>and</strong> 45 kg <strong>and</strong> aged 2 <strong>to</strong> 4 years. The endemic areas were thesame as for man. Hunting dogs generally cover large distances <strong>and</strong> can enterendemic areas with ecological niches of the fungus (Rudmann et al., 1992). Caseshave also been described in cats, a horse, a captive sea lion (Eume<strong>to</strong>pias jubata), anAfrican lion (Panthera leo) in a zoo, a dolphin, <strong>and</strong> a ferret. Cats follow dogs interms of numbers of cases, but the <strong>to</strong>tal number of cats affected is small. In a universityhospital in Tennessee (USA), 5 out of 5,477 cats treated presented blas<strong>to</strong>mycosis(Breider et al., 1988).The Disease in Man: The incubation period is not well known, but is estimated<strong>to</strong> be from 21 <strong>to</strong> 106 days (an average of 43 days) (Klein et al., 1986).Blas<strong>to</strong>mycosis may develop insidiously <strong>and</strong> silently, or acutely with symp<strong>to</strong>ms of afebrile disease, arthralgia, myalgia, <strong>and</strong> pleuritic pain. It may start with a dry coughthat becomes productive with hemoptysis, chest pain, <strong>and</strong> weight loss. Fever, cough,dyspnea, <strong>and</strong> diffuse pulmonary infiltration indicated by chest x-ray were seen in adescription of acute respira<strong>to</strong>ry distress syndrome in 10 adult patients. Six of thepatients had no underlying disease associated with a change in immunity <strong>and</strong> twohad no recent exposure <strong>to</strong> environmental reservoirs of B. dermatitidis. Microscopicexamination of tracheal secretions showed budding yeasts. Five of the 10 patientsdied, despite intravenous treatment with amphotericin B (Meyer et al., 1993).However, in most cases, the disease is asymp<strong>to</strong>matic at the outset <strong>and</strong> is diagnosedin a chronic state. The principal clinical form is pulmonary blas<strong>to</strong>mycosis. It is asystemic disease with a wide variety of pulmonary <strong>and</strong> extrapulmonary manifestations.The pulmonary form has the symp<strong>to</strong>ms of chronic pneumonia. The lesions aresimilar <strong>to</strong> those produced by other granuloma<strong>to</strong>us <strong>diseases</strong> (Chapman, 1990).The extrapulmonary forms are attributed <strong>to</strong> dissemination from the lungs. Thecutaneous form is the form most <strong>common</strong>ly seen in patients. Some patients do notpresent simultaneous pulmonary involvement. The disease is evidenced by verruci-


BLASTOMYCOSIS 313form lesions on exposed parts of the body or by an irregularly shaped scabby ulcerwith raised borders. A single patient may present both types of cutaneous lesions(Chapman, 1990). Other forms consist of subcutaneous nodules <strong>and</strong> particularlylesions in the joints, long bones, vertebrae, <strong>and</strong> ribs. The lesions are osteolytic <strong>and</strong>well defined, with abscesses forming in the soft tissue. A large number of patientsmay have prostate <strong>and</strong> epididymis lesions (Chapman, 1990).Of 15 AIDS patients in six endemic <strong>and</strong> four nonendemic areas, seven sufferedfrom a localized pulmonary blas<strong>to</strong>mycosis <strong>and</strong> eight from disseminated or extrapulmonaryblas<strong>to</strong>mycosis. Localization in the CNS was frequent (40% of cases). Six ofthe patients died in the first 21 days after admission <strong>to</strong> the medical facility with aclinical picture of blas<strong>to</strong>mycosis, two of them with fulminant pneumonia (Pappas etal., 1992). The authors conclude that blas<strong>to</strong>mycosis is a late <strong>and</strong> often fatal complicationthat occurs in a small number of AIDS patients.The preferred medication for disseminated cases is intravenous amphotericin B;ke<strong>to</strong>conazole is preferred for patients with more limited lesions, as it does not havethe amphotericin B side effects.The Disease in Animals: The highest incidence is seen in dogs around two yearsof age. Symp<strong>to</strong>ms consist of weight loss, chronic cough, dyspnea, cutaneousabscesses, fever, anorexia, <strong>and</strong>, with some frequency, blindness. The lesions localizein the lungs, lymph nodes, eyes, skin, <strong>and</strong> joints <strong>and</strong> bones. Of the 47 clinicalcases described, 72% occurred in large males. Lesions were present in the respira<strong>to</strong>rytract in 85% of the cases (Legendre et al., 1981). The number of cases in dogsis increasing in the United States; between January 1980 <strong>and</strong> July 1982, 200 casesof canine blas<strong>to</strong>mycosis were recorded in Wisconsin alone. Cases also have beenreported east of the Mississippi River (Archer et al., 1987). The preferred treatmentis the same as for man—intravenous amphotericin B. A large percentage of sickdogs are euthanized due <strong>to</strong> the high cost of treatment <strong>and</strong> the possible side effect ofnephro<strong>to</strong>xicity (Holt, 1980).Source of Infection <strong>and</strong> Mode of Transmission: The reservoir is environmental.Epidemiologic studies conducted in recent years reveal that the optimum microecosystemis s<strong>and</strong>y, acidic soil along waterways, <strong>and</strong> probably around artificialreservoirs as well (see the section on etiology). When environmental conditionschange, the agent isolated once often cannot be isolated again. Exposed men <strong>and</strong>dogs are those who come in<strong>to</strong> contact with the foci in endemic areas (see the sectionon geographic distribution), for work or recreation, particularly hunting.Transmission <strong>to</strong> man <strong>and</strong> animals is via the airborne route; fungal conidia are theinfecting element.Role of Animals in the Epidemiology of the Disease: None. It is a disease <strong>common</strong><strong>to</strong> man <strong>and</strong> animals. There are no known cases of transmission from one individual<strong>to</strong> another (man or animal).Diagnosis: Diagnosis is based on direct microscopic examination of sputum <strong>and</strong>material from lesions, on isolation of the agent in culture media, <strong>and</strong> on his<strong>to</strong>logicalpreparations. B. dermatitidis grows well in Sabouraud’s culture medium or anothersuitable culture medium. It is most distinctive in its budding yeast form, <strong>and</strong> thus theinoculated medium should be incubated at 37°C (the mycelial form of the fungus isobtained at ambient temperature). B. dermatitidis in its yeast form (in tissues or cul-


314 MYCOSEStures at 37°C) is characterized by a single bud attached <strong>to</strong> the parent cell by a widebase from which it detaches upon reaching a size similar <strong>to</strong> that of the parent cell.In contrast, Paracoccidioides brasiliensis, the agent of paracoccioidomycosis(“South American blas<strong>to</strong>mycosis”), has multiple buds in the yeast-forming phase. Acommercial DNA probe can be used <strong>to</strong> confirm the identity of the B. dermatitidisculture (Scalarone et al., 1992).Serological tests used are complement fixation <strong>and</strong> gel immunodiffusion; the latteryields the best results. Sensitivity is much greater in disseminated than in localizedblas<strong>to</strong>mycosis. An antigen-capture ELISA test proved <strong>to</strong> be more specific thanthe conventional enzymatic immunoassays: of eight serum samples obtained frompatients in an early stage of the disease, seven tested positive with this method,whereas three tested positive with gel immunodiffusion <strong>and</strong> three with complementfixation. There were no cross reactions with sera from patients with his<strong>to</strong>plasmosisor coccidioidomycosis (Lo <strong>and</strong> Notenboom, 1990), though it should be borne inmind that cross reactions with His<strong>to</strong>plasma <strong>and</strong> Coccidioides may occur. At present,the intradermal test is considered <strong>to</strong> have no diagnostic value. In dogs, serologicaltests have not yielded reliable results.Control: There are no adequate control measures.BibliographyAjello, L. Comparative ecology of respira<strong>to</strong>ry mycotic disease agents. Bact Rev 31:6–24, 1967.Archer, J.R., D.O. Trainer, R.F. Schell. Epidemiologic study of canine blas<strong>to</strong>mycosis inWisconsin. J Am Vet Med Assoc 190:1292–1295, 1987.Armstrong, C.W., S.R. Jenkins, L. Kaufman, et al. Common-source outbreak of blas<strong>to</strong>mycosisin hunters <strong>and</strong> their dogs. J Infect Dis 155:568–570, 1987.Bakerspigel, A., J.S. Kane, D. Schaus. Isolation of Blas<strong>to</strong>myces dermatitidis from anearthen floor in southwestern Ontario, Canada. J Clin Microbiol 24:890–891, 1986.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Breider, M.A., T.L. Walker, A.M. Legendre, R.T. VanEl. Blas<strong>to</strong>mycosis in cats: Five cases(1979–1986). J Am Vet Med Assoc 193:570–572, 1988.Chapman, S.W. Blas<strong>to</strong>myces dermatitidis. In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E.Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: ChurchillLivings<strong>to</strong>ne, Inc.; 1990.Drutz, D.J. The mycoses. In: Wyngaarden, J.B., L.H. Smith, Jr., eds. Cecil Textbook ofMedicine. 16th ed. Philadelphia: Saunders; 1982.Holt, R.J. Progress in antimycotic chemotherapy, 1945–1980. Infection 8:5284–5287, 1980.Kaplan, W. Epidemiology of the principal systemic mycoses of man <strong>and</strong> lower animals <strong>and</strong>the ecology of their agents. J Am Vet Med Assoc 163:1043–1047, 1973.Kaufman, L. Current status of immunology for diagnosis <strong>and</strong> prognostic evaluation of blas<strong>to</strong>mycosis,coccidioidomycosis, <strong>and</strong> paracoccidioidomycosis. In: Pan American HealthOrganization. Proceedings of the Third International Conference on the Mycoses.Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1975. (Scientific Publication 304).Kaufman, L., P.G. St<strong>and</strong>ard, R.J. Weeks, A.A. Padhye. Detection of two Blas<strong>to</strong>myces dermatitidisserotypes by exoantigen analysis. J Clin Microbiol 18:110–114, 1983.


CANDIDIASIS 315Klein, B.S., J.M. Vergeront, J.P. Davis. Epidemiologic aspects of blas<strong>to</strong>mycosis, the enigmaticsystemic mycosis. Semin Respir Infect 1:29–39, 1986.Legendre, A.M., M. Walker, N. Buyukmihci, R. Stevens. Canine blas<strong>to</strong>mycoses: A reviewof 47 clinical cases. J Am Vet Med Assoc 178:1163–1168, 1981.Lo, C.Y., R.H. Notenboom. A new enzyme immunoassay specific for blas<strong>to</strong>mycosis. AmRev Resp Dis 141:84–88, 1990.Lowry, P.W., K.Y. Kelso, L.M. McFarl<strong>and</strong>. Blas<strong>to</strong>mycosis in Washing<strong>to</strong>n Parish, Louisiana,1976–1985. Am J Epidemiol 130:151–159, 1989.Menges, R.W. Blas<strong>to</strong>mycosis in animals. Vet Med 55:45–54, 1960. Cited in: Selby, L.A.Blas<strong>to</strong>mycosis. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger, eds. DiseasesTransmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.Meyer, K.C., E.J. McManus, D.G. Maki. Overwhelming pulmonary blas<strong>to</strong>mycosis associatedwith the adult respira<strong>to</strong>ry distress syndrome. N Engl J Med 329:1231–1236, 1993.Pappas, P.G., J.C. Pottage, W.G. Powderly, et al. Blas<strong>to</strong>mycosis in patients with acquiredimmunodeficiency syndrome. Ann Intern Med 116:847–853, 1992.Rudmann, D.G., B.R. Coolman, C.M. Pérez, L.T. Glickman. Evaluation of risk fac<strong>to</strong>rs forblas<strong>to</strong>mycosis in dogs: 857 cases (1980–1990). J Am Vet Med Assoc 201:1754–1759, 1992.Scalarone, G.M., A.M. Legendre, K.A. Clark, K. Pusatev. Evaluation of a commercial DNAprobe assay for the identification of clinical isolates of Blas<strong>to</strong>myces dermatitidis from dogs. JMed Vet Mycol 30:43–49, 1992.Selby, L.A. Blas<strong>to</strong>mycosis. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger, eds.Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.CANDIDIASISICD-10 B37.0 c<strong>and</strong>idal s<strong>to</strong>matitis; B37.1 pulmonary c<strong>and</strong>idiasis;B37.2 c<strong>and</strong>idiasis of skin <strong>and</strong> nail; B37.3 c<strong>and</strong>idiasis of vulva <strong>and</strong> vagina;B37.4 c<strong>and</strong>idiasis of other urogenital sites; B37.5 c<strong>and</strong>idal meningitis;B37.6 c<strong>and</strong>idal endocarditis; B37.7 c<strong>and</strong>idal septicaemiaSynonyms: Moniliasis, c<strong>and</strong>idosis, thrush, c<strong>and</strong>idomycosis.Etiology: C<strong>and</strong>ida albicans (Monilia albicans, Oidium albicans) is the most <strong>common</strong>species in man <strong>and</strong> animals. Other less frequent species are C<strong>and</strong>ida tropicalis,C. parapsilosis, C. krusei, C. guillermondi, C. pseudotropicalis, <strong>and</strong> C. lusitaniae.C. albicans in young cultures measures approximately 3 x 5 microns. It is grampositive<strong>and</strong> reproduces by budding. In Sabouraud’s medium it forms creamy white,convex colonies. Old cultures have septate hyphae <strong>and</strong> sometimes chlamydospores(enlarged spherical cells with thick walls). C. albicans forms part of the normal florain the human <strong>and</strong> animal digestive system, mucosa <strong>and</strong>, <strong>to</strong> a lesser extent, the skin. Itis also found in the soil, in plants, <strong>and</strong> in fruits. In its normal habitat, C<strong>and</strong>ida takesthe form of a budding yeast. In infected tissue, it can produce hyphae or pseudohyphae(filaments consisting of elongated budding cells that did not detach from theparent cell). Odds <strong>and</strong> Abbott (1980, 1983) developed a biotyping method forC<strong>and</strong>ida albicans, later modified by Childress et al. (1989). This method consists of


316 MYCOSESevaluating the growth of the strain in nine agar plates with various biochemical compositionsin order <strong>to</strong> differentiate the strains for epidemiological purposes.Geographic Distribution: Worldwide. There are no delimited endemic zones.Occurrence in Man: It is the most frequent opportunistic mycosis. Its incidencehas increased in recent years due <strong>to</strong> the increase in prolonged treatments with antibiotics<strong>and</strong> corticosteroids. C<strong>and</strong>idiasis is a sporadic disease; epidemics have occurredin nurseries, particularly among premature babies in intensive care units; some epidemicsare due <strong>to</strong> the use of contaminated medicinal solutions or parenteral feedingfluids. It is estimated that the disease is responsible for nearly one-quarter ofmycotic deaths. In a general hospital in Mexico, c<strong>and</strong>idiasis lesions were found in5.4% of r<strong>and</strong>om au<strong>to</strong>psies conducted (González-Mendoza, 1970).Occurrence in Animals: The disease has been confirmed in numerousmammalian <strong>and</strong> avian species. Moniliasis in chicks <strong>and</strong> poults is <strong>common</strong> <strong>and</strong>sometimes has economic implications. Outbreaks have been described in variousparts of the world.The Disease in Man: C<strong>and</strong>ida is found as a commensal in the digestive tract <strong>and</strong>vagina of a high percentage of healthy individuals. Diaper rash <strong>and</strong> cheilitis (lipsores) are often caused by C<strong>and</strong>ida. In adults, c<strong>and</strong>idiasis is always associated withdebilitating <strong>diseases</strong> or conditions, such as diabetes (which particularly favorssuperficial c<strong>and</strong>idiasis), AIDS, tuberculosis, syphilis, cancer, obesity, <strong>and</strong> others.The agent often is responsible for intertrigo of large skin folds, balanitis, <strong>and</strong> onychiawith paronychia (especially in women whose work frequently requires them <strong>to</strong>immerse their h<strong>and</strong>s in water).The most frequent form of the mucosal infection presents clinically as a mycotics<strong>to</strong>matitis (thrush) characterized by lightly adhering white plaques on the <strong>to</strong>ngue<strong>and</strong> other parts of the mouth that can leave a bloody surface when removed. Somehave observed that this clinical form increased in asthmatic children treated withinhaled steroids. The infection often heals spontaneously (Edwards, 1990).The high incidence of thrush in cancer or AIDS patients should lead a physiciantreating a patient with thrush <strong>to</strong> test for these <strong>diseases</strong> (Syrjanen et al., 1988).Another form of mucosal infection is esophageal c<strong>and</strong>idiasis, which may or maynot be an extension of oral thrush. It is particularly frequent in patients receivingtreatment for malignant processes of the hema<strong>to</strong>poietic or lymphatic system. Themost <strong>common</strong> symp<strong>to</strong>ms of esophagitis are pain upon swallowing <strong>and</strong> substernalpain (Edwards, 1990).Gastrointestinal c<strong>and</strong>idiasis follows the esophageal form in frequency among cancerpatients. The small intestine is the third most frequent site of infection. Ulcersare the most <strong>common</strong> lesions in the s<strong>to</strong>mach <strong>and</strong> intestine.Mucosal c<strong>and</strong>idiasis recently has been surpassing Trichomonas as a cause of vulvovaginitis.This form is <strong>common</strong>ly accompanied by vaginal discharge of varyingintensity <strong>and</strong> pruritus vulvae.Although c<strong>and</strong>idiasis is usually limited <strong>to</strong> mucocutaneous forms, systemic infectioncan occur through hema<strong>to</strong>genous transmission, particularly in very weakpatients who are treated with antibiotics over a long period. These cases oftendevelop as a result of lesions caused by medical explorations using catheters, insertionof these instruments in the urethra, or surgical interventions. Though localiza-


CANDIDIASIS 317tion may occur in any organ, it is most frequent in the eyes, kidneys, lung, spleen,<strong>and</strong> the CNS, as well as around a cardiac value prosthesis or in the bones.The antimycotic recommended for mucosal <strong>and</strong> skin c<strong>and</strong>idiasis is nystatin; clotrimazoleis also effective. Amphotericin B or fluconazole are used <strong>to</strong> treat other sites.A cooperative study in 18 medical centers in Europe evaluated the efficacy,harmlessness, <strong>and</strong> <strong>to</strong>lerance of oral fluconazole (50 mg/day in a single dose) <strong>and</strong>of polyenes (oral amphotericin B at 2 g/day or nystatin at 4 million units/day infour or more doses) in preventing mycotic infection. The study included 536patients hospitalized with a malignant disease who were about <strong>to</strong> receivechemotherapy, radiotherapy, or bone marrow transplants, including patients whoalready had neutropenia or who were expected <strong>to</strong> develop it. Treatment was administeredfor approximately 30 days. Oral fluconazole proved <strong>to</strong> be more effectivethan the oral polyenes in preventing buccopharyngeal infection <strong>and</strong> was equallyeffective in preventing infections in other parts of the body in patients with neutropenia.Side effects were recorded in 5.6% of 269 patients in the group treatedwith fluconazole <strong>and</strong> 5.2% of 267 patients treated with polyenes. These reactionsled <strong>to</strong> a discontinuation of treatment in seven patients in each group (Philpott-Howard et al., 1993).The Disease in Animals: C<strong>and</strong>idiasis in chicks, poults, <strong>and</strong> other fowl is usuallysporadic. Epidemic outbreaks sometimes occur, particularly in poults, with mortalityranging from 8% <strong>to</strong> 20%. Avian c<strong>and</strong>idiasis is an infection of the upper respira<strong>to</strong>rysystem. In young birds it sometimes has an acute course, with nervous symp<strong>to</strong>ms.However, the disease is generally asymp<strong>to</strong>matic <strong>and</strong> diagnosis occurspostmortem. The most frequent lesion is found in the crop <strong>and</strong> consists of plaquesthat resemble curdled milk <strong>and</strong> adhere lightly <strong>to</strong> the mucosa. In adult birds, c<strong>and</strong>idiasishas a chronic course <strong>and</strong> causes thickening of the crop wall, on which a yellowishnecrotic material accumulates. In Israel, a strange epidemic of a venereal diseasein geese that affected many farms was described. The disease began withreddening <strong>and</strong> tumefaction of the mucosa of the penis or cloaca; the lesion laterbecame gangrenous <strong>and</strong> a portion of the penis was lost. Examinations indicated amixed flora of bacteria <strong>and</strong> C. albicans. Experimental inoculation of the bacterialflora did not affect the birds; in contrast, it was possible <strong>to</strong> reproduce the diseasewith C. albicans isolated from the lesions.Oral c<strong>and</strong>idiasis occurs sporadically in calves, colts, lambs, swine, dogs, cats, labora<strong>to</strong>rymice <strong>and</strong> guinea pigs, as well as in zoo animals. C<strong>and</strong>ida spp. can, on rareoccasions, lead <strong>to</strong> mastitis <strong>and</strong> abortions in cattle. A systemic disease due <strong>to</strong> C. albicans,with lesions in various organs, was reported in calves that underwent prolongedtreatment with antibiotics. Skin lesions <strong>and</strong> thrush have been described in cats.Source of Infection <strong>and</strong> Mode of Transmission: C. albicans occurs as a componen<strong>to</strong>f the normal flora in the digestive system of a high percentage of healthyindividuals <strong>and</strong> animals. The yeast is also found in nature.In young fowl, C. albicans is probably a primary etiologic agent, while in manc<strong>and</strong>idiasis is almost always associated with other <strong>diseases</strong>. Prolonged treatmentwith antibiotics, cy<strong>to</strong><strong>to</strong>xic agents, <strong>and</strong> corticosteroids is a predisposing fac<strong>to</strong>r. Theuse <strong>and</strong> abuse of antibiotics over an extended period is an important fac<strong>to</strong>r in theproliferation of <strong>and</strong> later infection by C<strong>and</strong>ida <strong>and</strong> other fungi, in that they alter thenatural flora of the mucosal surfaces.


318 MYCOSESMost infections have an endogenous source. The infection can be spread throughcontact with oral secretions, skin, vagina, <strong>and</strong> feces of sick individuals or carriers. Amother with vaginal c<strong>and</strong>idiasis can infect her child during childbirth. Balanitis mayin some cases be due <strong>to</strong> sexual relations with women suffering from vaginitis causedby C. albicans. In nurseries, particularly in units for premature infants, the infectionmay have an environmental source (see the section on occurrence in man). An exogenousinfection probably occurred due <strong>to</strong> indirect contact between patients in a hospitalbone marrow transplant unit <strong>and</strong> an intensive care unit (Vázquez et al., 1993).Role of Animals in the Epidemiology of the Disease: It is a disease <strong>common</strong> <strong>to</strong>man <strong>and</strong> animals. There are no known cases of transmission from animal <strong>to</strong> animal,but human-<strong>to</strong>-human transmission has occurred, as in the case of mothers who infecttheir children during childbirth.Diagnosis: Given the ubiqui<strong>to</strong>us nature of the yeast, labora<strong>to</strong>ry diagnosis must beconducted with great care. Direct examination of lesions in the nails, skin (in potassiumhydroxide) or the mucous membranes (in lac<strong>to</strong>phenol-cot<strong>to</strong>n blue), or microscopicobservation of gram-stained films, is diagnostically significant if the microorganismis found in great numbers. The examination should be carried out with freshspecimens. The presence in lesions of the budding yeast form <strong>to</strong>gether with formswith hyphae or pseudohyphae has diagnostic value. Isolation of the agent fromblood, pleural or peri<strong>to</strong>neal fluid, cerebrospinal fluid, or biopsy material obtainedaseptically from closed localized foci permits diagnosis of disseminated c<strong>and</strong>idiasis.However, it should be kept in mind that fungemia may be transient <strong>and</strong> is not alwaysindicative of systemic infection. Hemocultures can detect c<strong>and</strong>idemia in 35% <strong>to</strong>44% of patients with disseminated c<strong>and</strong>idiasis. C. albicans grows well in a mediumof blood agar <strong>and</strong> Sabouraud agar at 25°C <strong>and</strong> 37°C. It can be identified by demonstratingthe presence of chlamydospores upon seeding in depth a plate of corn mealagar <strong>and</strong> observing it at 24 <strong>and</strong> 48 hours. Since another characteristic of this speciesis the production of germinating tubes, identification can be performed by adding asmall amount of culture <strong>to</strong> a small amount of serum <strong>and</strong> incubating the mixture at37°C for two <strong>to</strong> four hours (Carter <strong>and</strong> Chengappa, 1991). The other species ofC<strong>and</strong>ida can be identified by their biochemical properties of carbohydrate fermentation<strong>and</strong> assimilation. A labeled anti-C. albicans globulin for immunofluorescencetesting of smears of pathologic or cultured materials is available.The most widely used serologic test <strong>to</strong> diagnose systemic c<strong>and</strong>idiasis is immunodiffusionor double diffusion in ouchterlony agar gel, which cumulative experiencehas shown <strong>to</strong> be highly sensitive <strong>and</strong> specific. The immunoelectrophoresis test correlateswell with the immunodiffusion test <strong>and</strong> results are obtained in only two hours.Nonetheless, serologic diagnosis of systemic c<strong>and</strong>idiasis presents serious difficulties<strong>and</strong> an increase in patients’ titers should be confirmed. Immunosuppressed patientshave a poor humoral response, <strong>and</strong> thus an attempt has been made <strong>to</strong> use techniquesthat detect antigenemia rather than circulating antibodies. To date the results have notbeen very encouraging. Sensitivity is low (Lemieux et al., 1990; Bougnoux et al.,1990). Tube agglutination, indirect immunofluorescence, <strong>and</strong> indirect hemagglutinationare also useful tests if the antibody level detected is above that prevalent in thenormal population. The predominant or sole antibodies in healthy individuals areIgM. In contrast, with systemic c<strong>and</strong>idiasis there is an initial rapid increase of IgM<strong>and</strong> then IgG, with subsequent reduction of IgM <strong>and</strong> persistence of IgG.


CANDIDIASIS 319Control: Neonatal c<strong>and</strong>idiasis can be prevented by treating the mother’s vaginalc<strong>and</strong>idiasis with nystatin during the third trimester. This antimycotic antibiotic canalso be used in patients undergoing prolonged treatment with broad-spectrum antibiotics.Plastic catheters should be avoided. Generalized thrush in weakened patientscan be halted by treating the oral lesions. To prevent epidemics in nurseries, patientswith oral thrush should be isolated <strong>and</strong> strict hygiene measures established. As a preventivemeasure, nutritional deficiencies should be corrected, given that c<strong>and</strong>idiasisoccurs with greater frequency in patients with vitamin deficiencies or inadequatediets (Ajello <strong>and</strong> Kaplan, 1980).Recommended control measures in case of a moniliasis outbreak among fowlinclude destroying all sick birds <strong>and</strong> administering copper sulphate (1:2,000) in thedrinking water <strong>and</strong> nystatin (110 mg/kg) in the feed.To date there is no vaccine.BibliographyAinsworth, G.C., P.K.C. Austwick. Fungal Diseases of Animals. 2nd ed. Farnham Royal,Slough, United Kingdom: Commonwealth Agricultural Bureau; 1973.Ajello, L., W. Kaplan. Systemic mycoses. In: S<strong>to</strong>enner, H., W. Kaplan, M. Torten, eds.Section A, Vol 2: CRC H<strong>and</strong>book Series in Zoonoses. Boca Ra<strong>to</strong>n: CRC Press; 1980.Anderson, K.L. Pathogenic yeasts. In: Hubbert, W.T., W.F. McCulloch, P.R.Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield:Thomas; 1975.Beemer, A.M., E.S. Kuttin, Z. Katz. Epidemic venereal disease due <strong>to</strong> C<strong>and</strong>ida albicans ingeese in Israel. Avian Dis 17:639–649, 1973.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bougnoux, M.E., C. Hill, D. Moissenet, et al. Comparison of antibody, antigen, <strong>and</strong>metabolite assays for hospitalized patients with disseminated or peripheral c<strong>and</strong>idiasis. J ClinMicrobiol 28:905–909, 1990.Carter, G.R. Diagnostic Procedures in Veterinary Microbiology. 2nd ed. Springfield:Thomas; 1973.Carter, G.R., M.N. Chengappa. Essentials of Veterinary Bacteriology <strong>and</strong> Mycology. 4th ed.Philadelphia: Lea & Febiger; 1991.Childress, C.M., J.A. Holder, A.N. Neely. Modifications of a C<strong>and</strong>ida albicans biotypingsystem. J Clin Microbiol 27:1392–1394, 1989.Edwards, J.E., Jr. C<strong>and</strong>ida species. In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E. Bennett, eds.Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: Churchill Livings<strong>to</strong>ne,Inc.; 1990.González-Mendoza, A. Opportunistic mycoses. In: Pan American Health Organization.Proceedings: International Symposium on Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1970.(Scientific Publication 205).Gordon, M.A. Current status of serology for diagnosis <strong>and</strong> prognostic evaluation of opportunisticfungus infections. In: Pan American Health Organization. Proceedings of the ThirdInternational Conference on the Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1975. (ScientificPublication 304).Lemieux, C., G. St.-Germain, J. Vincelette, et al. Collaborative evaluation of antigen detectionby a commercial latex agglutination test <strong>and</strong> enzyme immunoassay in the diagnosis ofinvasive c<strong>and</strong>idiasis. J Clin Microbiol 28:249–253, 1990.


320 MYCOSESNegroni, P. Micosis cutáneas y viscerales. 5.ª ed. Buenos Aires: López; 1972.Odds, F.C., A.B. Abbott. A simple system for the presumptive identification of C<strong>and</strong>idaalbicans <strong>and</strong> differentiation of strains within the species. Sabouraudia 18:301–317, 1980.Odds, F.C., A.B. Abbott. Modification <strong>and</strong> extension of tests for differentiation of C<strong>and</strong>idaspecies <strong>and</strong> strains. Sabouraudia 21:79–81, 1983.Philpott-Howard, J.N., J.J. Wade, G.J. Mufti, et al. R<strong>and</strong>omized comparison of oral flucanozoleversus oral polyenes for the prevention of fungal infection in patients at risk of neutropenia.Multicentre Study Group. J Antimicrob Chemother 31:973–984, 1993.Soltys, M.A. Bacteria <strong>and</strong> Fungi Pathogenic <strong>to</strong> Man <strong>and</strong> Animals. London: Baillière-Tindall; 1963.Syrjanen, S., S.L. Valle, J. An<strong>to</strong>nen, et al. Oral c<strong>and</strong>idal infection as a sign of HIV infectionin homosexual men. Oral Surg Oral Med Oral Pathol 65:36–40, 1988.Vázquez, J.A., V. Sánchez, C. Dmuchowski, et al. Nosocomial acquisition of C<strong>and</strong>ida albicans:An epidemiologic study. J Infect Dis 168:195–201, 1993.COCCIDIOIDOMYCOSISICD-10 B38.0 acute pulmonary coccidioidomycosis; B38.1 chronic pulmonarycoccidioidomycosis; B38.3 cutaneous coccidioidomycosis; B38.7 disseminatedcoccidioidomycosis; B38.8 other forms of coccidioidomycosisSynonyms: Posada’s disease, San Joaquin Valley fever, desert fever.Etiology: Coccidioides immitis,a diphasic fungus that exists in the mycelial phasewhen it is a soil saprophyte, <strong>and</strong> in the spherule phase in organic tissues <strong>and</strong> fluids.The life cycle of C. immitis is unique among pathogenic fungi. The fungus occursin one phase in the natural environment, i.e., the soil of semiarid regions, <strong>and</strong> inanother when it is parasitic in the mammalian host. In the soil, C. immitis developsas a mycelium (a mass of filamen<strong>to</strong>us hyphae that make up the fungus). The cyclebegins with the arthroconidium, or arthrospore (spore formed in the hyphae), whichin a suitable medium germinates <strong>and</strong> forms a branching, septate mycelium. Whenthe mycelium fragments, it releases in<strong>to</strong> the air arthroconidia 2 <strong>to</strong> 5 microns in size.The parasitic phase begins with the inhalation of arthroconidia by man <strong>and</strong> animals.Arthroconidia grow <strong>to</strong> form thick-walled spherules 10 <strong>to</strong> 80 microns in diameter.The cy<strong>to</strong>plasm of the spherules divides <strong>to</strong> produce hundreds of endospores which,when released, disperse in<strong>to</strong> the surrounding tissue <strong>and</strong> give rise <strong>to</strong> new spherules.The parasitic cycle lasts from four <strong>to</strong> six days (Drutz <strong>and</strong> Huppert, 1983) <strong>and</strong> canrevert <strong>to</strong> the saprophytic or mycelial phase if the endospores reach the soil upon thedeath of the infected animal or through bodily excretions. The endospores give rise<strong>to</strong> hyphae <strong>and</strong> renew the cycle (Stevens, 1990). However, the mycelial cycle doesnot depend on this reversion as the hyphae contain large amounts of arthroconidiathat are dispersed by the wind <strong>and</strong> colonize new sites in the soil.Geographic Distribution: Limited <strong>to</strong> the Americas. The fungus is found in arid<strong>and</strong> semiarid areas of the United States Southwest, northwestern Mexico, Argentina,


COCCIDIOIDOMYCOSIS 321Colombia, Guatemala, Honduras, Paraguay, Venezuela, <strong>and</strong> probably Bolivia. Theendemic area in Latin America is estimated <strong>to</strong> cover 1.5 million km 2 , more than1 million km 2 of which are in Mexico (Borelli, 1970).Occurrence in Man: In some endemic areas the rate of infection seems <strong>to</strong> bevery high <strong>and</strong> it is estimated that in some of these areas in the United States nearly100% of the population could contract the infection within a few years (Fiese, citedby Ajello, 1970). There are an estimated 25,000 <strong>to</strong> 100,000 cases in the US eachyear. Approximately 20% of the cases involve people who live outside endemicareas <strong>and</strong> become infected while visiting them (Drutz <strong>and</strong> Huppert, 1983). Somecases have also been described in Europe (the former Czechoslovakia, Great Britain,<strong>and</strong> Denmark). The rate of reac<strong>to</strong>rs <strong>to</strong> the skin test in different endemic areas variesfrom 5% <strong>to</strong> more than 50% of the population. There is a significant increase in casesin the United States. In 1991, 1,208 new cases were recorded in California as compared<strong>to</strong> 450 cases per year on average in the previous five years. Of these cases,80% came from Kern County, a known endemic area. Sixty-three percent of thecases were reported from Oc<strong>to</strong>ber through December. The outbreak in Californiacould have been associated with prolonged drought, followed by occasional heavyrains. Another important fac<strong>to</strong>r could be migration <strong>to</strong> California of people not previouslyexposed <strong>to</strong> the fungus. In the United States, endemic areas are found inArizona, California, Nevada, New Mexico, Texas, <strong>and</strong> Utah (CDC, 1993). The dataon South America are more fragmentary, but the rate of infection appears <strong>to</strong> be lowerin this region.Occurrence in Animals: Natural infection has been found in many species ofmammals. Infection is very frequent in cattle <strong>and</strong> dogs in endemic areas. Veterinaryinspection has discovered coccidioidomycosis lesions in 5% <strong>to</strong> 15% of the cattleslaughtered in abat<strong>to</strong>irs in central Arizona (USA). Several million cattle are thought<strong>to</strong> be infected in the endemic areas of the southwestern United States. Infection hasalso been demonstrated in sheep, horses, swine, <strong>and</strong> wild rodents.Several studies were carried out on animals in the endemic region of Mexico. Inthe state of Sinaloa, sera from 100 hogs <strong>and</strong> 200 cattle were examined by immunoelectrophoresis<strong>and</strong> reactions were found in 12% <strong>and</strong> 13%, respectively (VelascoCastrejón <strong>and</strong> Campos Nie<strong>to</strong>, 1979). In the state of Sonora, when the intradermaltest using coccidioidin was conducted on 459 cattle, 6.75% tested positive. Anotherstudy performed his<strong>to</strong>logical examinations of granuloma<strong>to</strong>us lesions discovered in3,032 slaughtered cattle <strong>and</strong> found that the lesions in 77 (44%) of 175 animals confiscatedfor suspected tuberculosis were actually caused by C. immitis, indicating arate of infection of 2.5% in all the animals (Cervantes et al., 1978).The Disease in Man: The incubation period lasts from one <strong>to</strong> four weeks. Anestimated 60% of infections occur asymp<strong>to</strong>matically <strong>and</strong> are only recognizable withthe intradermal test. The remaining 40% present as a respira<strong>to</strong>ry disease with acutesymp<strong>to</strong>ms similar <strong>to</strong> those of influenza <strong>and</strong> that generally pass without sequelae.About 5% of primary infections develop either an erythema multiforme or an erythemanodosum arthralgia. What is more <strong>common</strong>, however, is a light erythrodermaor maculopapular eruption. Chest pain can be strong <strong>and</strong> pleuritic. The radiologicalpicture is varied, but hilar adenopathy with alveolar infiltrates <strong>and</strong> infiltrates thatchange area are indicative of coccidioidal pneumonia (Ampel et al., 1989). When


322 MYCOSESthe primary respira<strong>to</strong>ry disease does have sequelae, these consist of fibrotic or cavernouslesions in the lungs. Pneumonia may persist in some patients for six <strong>to</strong> eightweeks, accompanied by fever, chest pain, cough, or postration (persistent coccidioidalpneumonia). Mortality in these cases is high in immunocompromisedpatients. Another disease form is the chronic form, which can be confused withtuberculosis (Drutz, 1982).Extrapulmonary dissemination generally occurs following the primary disease inapproximately 0.5% of infections (CDC, 1993). Thoracic radiography may or maynot show abnormalities. The most <strong>common</strong> localization is in the cutaneous <strong>and</strong> subcutaneoustissues. Cutaneous lesions generally consist of verruciform granulomas(usually on the face), erythroma<strong>to</strong>us plaques, <strong>and</strong> nodules. Sometimes there are subcutaneousabscesses. Osteomyelitis occurs in 10% <strong>to</strong> 50% of disseminated cases <strong>and</strong>may affect one or more bones. Meningitis cases are frequent (33% <strong>to</strong> 50% ofpatients) <strong>and</strong> generally fatal within two years. Eosinophilic pleocy<strong>to</strong>sis is frequentin coccidioidal meningitis <strong>and</strong> has diagnostic value (Ragl<strong>and</strong> et al., 1993). Othermanifestations are thyroiditis, tenosynovitis, <strong>and</strong> prostatis (Drutz, 1982). Clinicalcoccidioidomycosis is more frequent among migrant workers <strong>and</strong> soldiers transferred<strong>to</strong> endemic zones. In endemic areas of C. immitis the symp<strong>to</strong>matic form ofthe disease is frequent in individuals infected by the human immunodeficiencyvirus. Immunodeficiency is an important risk fac<strong>to</strong>r for developing the disease(Ampel et al., 1993).Treatment is difficult <strong>and</strong> often unpredictable. Fungicides that were effective insome cases were not in other similar cases. It is estimated that less than 5% of thoseinfected need treatment. Those who are suffering from a progressive illness, patientswith severe primary pulmonary disease, <strong>and</strong> those who have disseminated infectionshould be treated. Treatment should also be considered for patients with a compromisedimmune system. Amphotericin B <strong>and</strong> ke<strong>to</strong>conazole are the medications mostfrequently used (Ampel et al., 1989). The administration of 400 mg of fluconazoledaily for up <strong>to</strong> four years <strong>to</strong> 47 patients with coccidioidal meningitis produced afavorable result in 37 patients (Galgiani et al., 1993).The Disease in Animals: The infection is asymp<strong>to</strong>matic in cattle. Lesions aregenerally limited <strong>to</strong> the bronchial <strong>and</strong> mediastinal lymph nodes. On rare occasions,small granuloma<strong>to</strong>us lesions are found in the lungs <strong>and</strong> the submaxillary <strong>and</strong>retropharyngeal lymph nodes. Macroscopic lesions resemble those seen in cases oftuberculosis.Ziemer et al. (1992) conducted a retrospective study of 15 cases of coccidioidomycosisin horses recorded from 1975 <strong>to</strong> 1984 in a university hospital inCalifornia, with diagnosis confirmed by culture or his<strong>to</strong>pathology. The most <strong>common</strong>symp<strong>to</strong>m in 53% of the horses was chronic weight loss, which ranged from45.5 kg <strong>to</strong> 91 kg in three horses. One of the horses lost 24% of its body weight inthree months. Thirty-three percent of the horses had a persistent cough. Sixty percen<strong>to</strong>f the animals had respira<strong>to</strong>ry abnormalities detected through auscultation.Other symp<strong>to</strong>ms were depression <strong>and</strong> superficial abscesses.Various cases have been described in sheep, with lesions similar <strong>to</strong> those in cattle.In the same university hospital in California, 19 cases of coccidioidomycosis wererecorded in llamas (10 from Arizona <strong>and</strong> 9 from California). Eighteen of the animalshad disseminated mycosis, with pyogranulomas in the lungs, thoracic ganglia, liver,


COCCIDIOIDOMYCOSIS 323<strong>and</strong> kidneys. The llama seems <strong>to</strong> be highly susceptible <strong>to</strong> infection by C. immitis. Itis not known whether there are unapparent or slight infections in this species(Fowler et al., 1992).After man, the dog is the species most affected. In addition <strong>to</strong> the lungs, granuloma<strong>to</strong>uslesions are found in nearly all organs. The disseminated form of the diseaseis frequent in dogs <strong>and</strong> the disease advances progressively until death (Timoney etal., 1988).Source of Infection <strong>and</strong> Mode of Transmission: C. immitis is a soil saprophytein arid <strong>and</strong> semiarid regions. Its distribution in endemic zones is not uniform. Theinfection is transmitted <strong>to</strong> man <strong>and</strong> animals through inhalation of wind-bornearthrospores of the fungus; it occurs more frequently after dust s<strong>to</strong>rms. The infectioncan be contracted in the labora<strong>to</strong>ry by inhaling the spores from fungus cultures.Exposure <strong>to</strong> soil with a high concentration of the agent increases the risk of asymp<strong>to</strong>matic <strong>and</strong> severe disease. This was probably the case with two archeologystudents on a dig in southern California (Larsen et al., 1985; Ampel et al., 1989).Those most exposed <strong>to</strong> contracting the infection are individuals without a his<strong>to</strong>ryof the infection who visit or migrate <strong>to</strong> endemic areas.Coccidioidomycosis is currently increasing in the United States due <strong>to</strong> significantgrowth in population <strong>and</strong> <strong>to</strong>urism in endemic areas.In recent decades, due <strong>to</strong> the great increase in the use of immunosuppressantdrugs for transplants, oncology, <strong>and</strong> rheuma<strong>to</strong>logy, as well as <strong>to</strong> AIDS, the severeform of the disease is seen more frequently (Ampel et al., 1989).Role of Animals: The soil is the <strong>common</strong> source of infection for man <strong>and</strong> animals.The fungus is not transmitted from one individual <strong>to</strong> another, because man <strong>and</strong>other infected animals do not produce arthroconidia, the infecting agent. An exceptionalcase due <strong>to</strong> aerosolization of endospores occurred during the au<strong>to</strong>psy of ahorse with disseminated coccidioidomycosis. The veterinarian who performed theau<strong>to</strong>psy contracted the infection by inhaling the endospores (Kohn et al., 1992).Diagnosis: Diagnosis is based on confirmation of the fungus’s presence by meansof: (1) direct microscopic examination that reveals spherules with endospores insputum, pus, pleural fluid, or gastric juices (treated with a 10% solution of potassiumhydroxide); (2) culture of clinical material; <strong>and</strong> (3) his<strong>to</strong>pathology. Culturesshould not be prepared in Petri dishes but in closed tubes so as <strong>to</strong> avoid infection ofthe h<strong>and</strong>ler <strong>and</strong> labora<strong>to</strong>ry personnel. Appropriate biosafety equipment should alsobe used.The skin test using coccidioidin or spherulin (considered <strong>to</strong> be more sensitive) isvery valuable in epidemiologic studies. It is administered in the same way as tuberculin.The test should be read at 24 <strong>and</strong> 48 hours. A reaction of 5 mm or more is consideredpositive. This test is very useful for delimiting endemic areas. In infectionsby C. immitis there may be cross-reactions with other fungal antigens, especiallyhis<strong>to</strong>plasmin. In clinical diagnosis, the intradermal test with a positive result is onlysignificant if the patient had no reaction at the beginning of the illness. In a studycomparing the tests with coccidioidin (prepared from the mycelial phase fungus)<strong>and</strong> spherulin (parasitic phase fungus) in patients with coccidioidomycosis, onepreparation could not be shown superior <strong>to</strong> the other for diagnosis. Forty-three percen<strong>to</strong>f the patients reacted positively <strong>to</strong> both preparations, another 43% reacted


324 MYCOSESnegatively <strong>to</strong> both, <strong>and</strong> 14% has contradic<strong>to</strong>ry results. The lack of reaction in a highpercentage of patients is perhaps due <strong>to</strong> defects in immune function, particularly inthe case of advanced disease (Gifford <strong>and</strong> Catanzaro, 1981). Serologic tests in useare complement fixation (CF), precipitation, immunodiffusion, <strong>and</strong> latex agglutination.The combination of immunobiological tests provides useful information forboth diagnosis <strong>and</strong> prognosis. In the first two weeks of the disease, IgM antibodiespredominate, as can be demonstrated by the tube precipitation, latex agglutination,<strong>and</strong> immunodiffusion tests. IgG antibodies appear somewhat later <strong>and</strong> may bedetected through CF or immunodiffusion. A persistent high CF titer with loss ofreactivity <strong>to</strong> the skin test indicates dissemination of the infection. In 75% <strong>to</strong> 95% ofmeningitis cases, antibodies can be detected with the CF test (Drutz, 1982).Radioimmunoassay is useful for diagnosis <strong>and</strong> prognosis of the pulmonary disease.As patients improve, the test titer decreases (Catanzaro <strong>and</strong> Flataner, 1983). The CFtest also indicates the efficacy of the treatment.Control: It is recommended that persons from nonendemic areas not work inendemic areas, since they lack immunity against coccidioidomycosis. In the UnitedStates, dust control measures (paving roads, seeding lawns, sprinkling dust with oil)have been used successfully <strong>to</strong> protect military personnel.People at risk of contracting disseminated coccidioidomycosis (pregnant women,immunocompromised patients) should be advised <strong>to</strong> avoid endemic areas. Trials ofa vaccine made from formalin-inactivated spherules are being conducted inCalifornia <strong>and</strong> Arizona (USA). Animal tests have shown that the vaccine does notprevent the infection, but does arrest its progress <strong>and</strong> prevent dissemination of thedisease (Drutz <strong>and</strong> Huppert, 1983). A test conducted from 1980 <strong>to</strong> 1985 with 1,436vaccinated subjects <strong>and</strong> 1,431 subjects given a placebo showed a slight but statisticallyinsignificant reduction in the incidence of coccidioidomycosis in the vaccinatedgroup as compared <strong>to</strong> the group receiving the placebo. There was no differencebetween the two groups in the severity of the disease (Pappagianis, 1993).Treatment with antifungal drugs may be useful <strong>to</strong> prevent dissemination in high-riskpatients with primary coccidioidomycosis.BibliographyAjello, L. Comparative ecology of respira<strong>to</strong>ry mycotic disease agents. Bact Rev 31:6–24, 1967.Ajello, L. The medical mycological iceberg. In: Pan American Health Organization.Proceedings: International Symposium on Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1970.(Scientific Publication 205).Ajello, L., L.K. Georg, W. Kaplan, L. Kaufman. Labora<strong>to</strong>ry Manual for MedicalMycology. Washing<strong>to</strong>n, D.C.: U.S. Government Printing Office; 1963. (Public HealthService Publication 994).Ampel, N.M., C.L. Dols, J.N. Galgiani. Coccidioidomycosis during human immunodeficiencyvirus infection: Results of prospective study in a coccidioidal endemic area. Am J Med94:235–240, 1993.Ampel, N.M., M.A. Wieden, J.N. Galgiani. Coccidioidomycosis: Clinical update. RevInfect Dis 11:897–911, 1989.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.


COCCIDIOIDOMYCOSIS 325Borelli, D. Prevalence of systemic mycoses in Latin America. In: Pan American HealthOrganization. Proceedings: International Symposium on Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>;1970. (Scientific Publication 205).Catanzaro, A., F. Flataner. Detection of serum antibodies in coccidioidomycosis by solidphaseradioimmunoassay. J Infect Dis 147:32–39, 1983.Cervantes, R.A., A.J. Solózano, C.B.J. Pijoan. Presencia de coccidioidomicosis en bovinosdel Estado de Sonora. Rev Latinoam Microbiol 20:247–249, 1978.Davis, J.W. Coccidioidomycosis. In: Davis, J.W., L.H. Karstad, D.O. Trainer, eds.Infectious Diseases of Wild Mammals. Ames: Iowa State University Press; 1970.Drutz, D.J. The mycoses. In: Wyngaarden, J.B., L.H. Smith, Jr., eds. Cecil Textbook ofMedicine. 16th ed. Philadelphia: Saunders; 1982.Drutz, D.J., M. Huppert. Coccidioidomycosis: Fac<strong>to</strong>rs affecting the host-parasite interaction.J Infect Dis 147:372–390, 1983.Fiese, M.J. Coccidioidomycosis. Springfield: Thomas; 1958. Cited in: Ajello, L. The medicalmycological iceberg. In: Pan American Health Organization. Proceedings: InternationalSymposium on Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1970. (Scientific Publication 205).Fowler, M.E., D. Pappagianis, I. Ingram. Coccidioidomycosis in llamas in the UnitedStates: 19 cases (1981–1989). J Am Vet Med Assoc 201:1609–1614, 1992.Galgiani, J.N., A. Catanzaro, G.A. Cloud, et al. Fluconazole therapy for coccidioidalmeningitis. The NIAID-Mycoses Study Group. Ann Intern Med 119:28–35, 1993.Gifford, J., A. Catanzaro. A comparison of coccidioidin <strong>and</strong> spherulin skin testing in thediagnosis of coccidioidomycosis. Am Rev Resp Dis 124:440–444, 1981.Kohn, G.J., S.R. Linne, C.M. Smith, P.D. Hoeprich. Acquisition of coccidioidomycosis atnecropsy by inhalation of coccidioidal endospores. Diagn Microbiol Infect Dis 15:527–530,1992.Larsen, R.A., J.A. Jacobson, A.H. Morris, B.A. Benowitz. Acute respira<strong>to</strong>ry failure causedby primary pulmonary coccidioidomycosis. Two case reports <strong>and</strong> a review of the literature.Am Rev Resp Dis 131:797–799, 1985.Maddy, K.T. Coccidioidomycosis. Adv Vet Sci 6:251–286, 1960.Negroni, K.T. Micosis cutáneas y viscerales. 5.ª ed. Buenos Aires: López; 1972.Pappagianis, D. Evaluation of protective efficacy of the killed Coccidioides immitisspherule vaccine in humans. The Valley Fever Vaccine Study Group. Am Rev Resp Dis148:656–660, 1993.Ragl<strong>and</strong>, A.S., E. Arsura, Y. Ismail, R. Johnson. Eosinophilic pleocy<strong>to</strong>sis in coccidioidalmeningitis: Frequency <strong>and</strong> significance. Am J Med 95:254–257, 1993.Stevens, D.A. Coccidioides immitis. In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E. Bennett, eds.Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: Churchill Livings<strong>to</strong>ne,Inc.; 1990.Timoney, J.F., J.H. Gillespie, F.W. Scott, J.E. Barlough. Hagan <strong>and</strong> Bruner’s Microbiology<strong>and</strong> Infectious Diseases of Domestic Animals. 8th ed. Ithaca: Coms<strong>to</strong>ck; 1988.United States of America, Department of Health <strong>and</strong> Human Services, Centers for DiseaseControl <strong>and</strong> Prevention (CDC). Coccidioidomycosis—United States, 1991–1992. MMWRMorb Mortal Wkly Rep 42:21–24, 1993.Velasco Castrejón, O., E. Campos Nie<strong>to</strong>. Estudio serológico de la coccidioidomicosis bovinay porcina del Estado de Sinaloa (México). Rev Latinoam Microbiol 21:99, 1979.Ziemer, E.L., D. Pappagianis, J.E. Madigan, et al. Coccidioidomycosis in horses: 15 cases(1975–1984). J Am Vet Med Assoc 201:910–916, 1992.


326 MYCOSESCRYPTOCOCCOSISICD-10 B45.0 pulmonary cryp<strong>to</strong>coccosis; B45.1 cerebral cryp<strong>to</strong>coccosis;B45.2 cutaneous cryp<strong>to</strong>coccosis; B45.3 osseous cryp<strong>to</strong>coccosis;B45.7 disseminated cryp<strong>to</strong>coccosis; B45.8 other forms of cryp<strong>to</strong>coccosisSynonyms: Torulosis, European blas<strong>to</strong>mycosis, Busse-Buschke’s disease.Etiology: Cryp<strong>to</strong>coccus neoformans (Saccharomyces neoformans, Torulopsisneoformans, Torula his<strong>to</strong>lytica), a saprophytic yeast growing in certain soils. Theagent has a spheroid or ovoid shape, is encapsulated, ranges from 4 <strong>to</strong> 7 microns indiameter, <strong>and</strong> is gram-positive. It reproduces by means of buds attached by a delicatebase <strong>to</strong> the parent cell. Research in recent years has demonstrated that C. neoformanshas a sexual form <strong>and</strong> is a basidiomycete.Of epidemiological interest is C. neoforman’s subdivision in<strong>to</strong> four serotypes (A,B, C, <strong>and</strong> D) on the basis of capsular polysaccharide antigens. In turn, the serotypesare categorized in two varieties: C. neoformans var. neoformans (serotypes A <strong>and</strong> D)<strong>and</strong> C. neoformans var. gattii (serotypes B <strong>and</strong> C). In addition <strong>to</strong> biochemical, serological,<strong>and</strong> genetic differences, serotypes A <strong>and</strong> D are different in their perfect (sexual)state from serotypes B <strong>and</strong> C. Although a few A <strong>and</strong> D strains can be conjugatedwith B <strong>and</strong> C, their survival is short-lived (Diamond, 1990).Geographic Distribution: Worldwide. In the Americas the disease has been confirmedin Argentina, Brazil, Canada, Colombia, Mexico, the United States, <strong>and</strong>Venezuela. Serotype A is prevalent throughout the world. Serotype D is <strong>common</strong> insome European countries (Denmark, Italy, <strong>and</strong> Switzerl<strong>and</strong>), but rare in the UnitedStates. In contrast, serotypes B <strong>and</strong> C are more localized <strong>and</strong> are recognized as diseaseagents, particularly in southern California, southeastern Oklahoma, <strong>and</strong> some otherareas of the United States, as well as in Asia (Kaplan et al., 1981; Fromtling et al.,1982). In some regions of Australia, a high percentage of isolated strains have thecharacteristics of the gattii strain, as in the case of the indigenous population in theNorthern Terri<strong>to</strong>ry. In one study, 25 of 26 isolates (24 of them from meningitispatients) corresponded <strong>to</strong> gattii. In another study, 21 of 22 strains (95.5%) were alsoof the gattii variety. In South Australia, which has a primarily urban population, 65.2%of 23 strains were classified as gattii (Ellis, 1987). Other sites with a high prevalenceof gattii var. are Brazil (10 of 31 strains) <strong>and</strong> southern California (30 of 73) (Kwon-Chung <strong>and</strong> Bennett, 1984). In Argentina, 101 of 105 isolates from 1981–1990 wereclassified as C. neoformans var. neoformans <strong>and</strong> 4 as var. gattii (serotype B). Thesedata are similar <strong>to</strong> those found in the United States (Bava <strong>and</strong> Negroni, 1992).Occurrence in Man: Cases are sporadic, with a higher incidence in men than inwomen. From 1965 <strong>to</strong> 1997, 1,264 cases of cryp<strong>to</strong>coccosis were documented in theUnited States. Of 848 cases confirmed between 1973 <strong>and</strong> 1997, 608 patients hadmeningitis <strong>and</strong> 240 had extrameningeal localizations. These data indicate a greatincrease as compared <strong>to</strong> earlier periods (Kaufman <strong>and</strong> Blumer, 1978). There were85 cases in Malaysia between 1974 <strong>and</strong> 1980, predominantly among ethnic Chinese(Pathmanathan <strong>and</strong> Soo-Hoo, 1982). In the United States <strong>and</strong> Europe, cryp<strong>to</strong>coccosisoccurs primarily in patients with immune system defects (especially AIDS) orwho are undergoing immunosuppressant treatment. The prevalence of the disease


CRYPTOCOCCOSIS 327has grown worldwide as the number of AIDS patients has increased. In Argentina,the annual number of cases ranged from four <strong>to</strong> eight until 1987, began <strong>to</strong> increasein 1988, <strong>and</strong> reached 35 cases in 1990. The age group most affected was 20- <strong>to</strong> 39-year-olds. More men were affected than women, particularly when the underlyingdisease was AIDS (Bava <strong>and</strong> Negroni, 1992). It is estimated that the male-femaleratio was 3:1. The percentage of AIDS patients who contracted cryp<strong>to</strong>coccosis inArgentina increased from 12.5% in 1990 <strong>to</strong> 25.9% in 1991. This percentage is similar<strong>to</strong> the incidence of cryp<strong>to</strong>coccosis in AIDS patients in central Africa <strong>and</strong> southeastAsia (20% <strong>to</strong> 35%), but greater than that in Europe <strong>and</strong> the United States (6%<strong>to</strong> 10%). In greater Buenos Aires, cryp<strong>to</strong>coccosis is second among the tracer <strong>diseases</strong>of AIDS, after esophageal c<strong>and</strong>idiasis (Bava et al., 1992). In Malaysia, on theother h<strong>and</strong>, only 14% of the patients studied had AIDS.Epidemiologic studies based on the intradermal test indicate that many individualsexposed <strong>to</strong> the agent show no symp<strong>to</strong>ms of the disease.Occurrence in Animals: Rare, sporadic cases. Some epizootic outbreaks of mastitis<strong>and</strong> cryp<strong>to</strong>coccal pneumonia have been described in cattle. The disease has beendescribed in goats, horses, <strong>and</strong> cats.The Disease in Man: The large majority of cases are meningitis or meningoencephalitis.This form is preceded by a pulmonary infection, which is often asymp<strong>to</strong>maticor, if symp<strong>to</strong>matic, may resolve spontaneously. In most cases of localizationin the CNS, pulmonary invasion is not evident (Diamond, 1990). The initial pulmonaryinfection can resolve spontaneously, give rise <strong>to</strong> a granuloma<strong>to</strong>us mass(“cryp<strong>to</strong>coccoma”), or disseminate via the bloodstream. The pulmonary form manifestswith fever, cough, chest pain, <strong>and</strong> hemoptysis. Radiography shows single ormultiple nodules or large cryp<strong>to</strong>coccomas. The course is usually chronic. When disseminationfrom the original pulmonary focus occurs, the infection localizes primarilyin the meninges, spreading <strong>to</strong> the brain. The most obvious symp<strong>to</strong>ms of themeningeal form of the disease are headache <strong>and</strong> visual disturbances. Other symp<strong>to</strong>msmay include confusion, personality changes, agitation, <strong>and</strong> lethargy.Cryp<strong>to</strong>coccal meningoencephalitis can follow a course lasting for weeks or months<strong>and</strong> is almost always fatal if not properly treated. The characteristic lesion in thebrain is comprised of groups of fungal cysts without inflammation. This lesion canalso be found in other sites (Diamond, 1990). Asymp<strong>to</strong>matic meningitis sometimesoccurs when there are other locations <strong>and</strong> the disease is discovered through lumbarpuncture <strong>and</strong> culture of the cerebrospinal fluid (Liss <strong>and</strong> Riml<strong>and</strong>, 1981). The lesioncan affect the skin, the mucosa, <strong>and</strong> the bones, as well as various other organs.Cutaneous infection is characterized by the formation of papules <strong>and</strong> abscesses <strong>and</strong>subsequent ulceration.Man is resistant <strong>to</strong> C. neoformans. There are cryp<strong>to</strong>coccosis patients who show noobvious predisposing fac<strong>to</strong>rs. However, the fungus is <strong>to</strong> a large extent an opportunisticpathogenic agent. The number of cases increased significantly with the HIVepidemic. In the United States, cryp<strong>to</strong>coccosis is the fourth potential leading case ofdeath in AIDS patients, after Pneumocystis carinii, cy<strong>to</strong>megaloviruses, <strong>and</strong>mycobacteria. A retrospective study of AIDS patients was conducted in a hospital inPor<strong>to</strong> Alegre, Brazil <strong>to</strong> determine the <strong>diseases</strong> that could affect the CNS. Between1985 <strong>and</strong> 1990, 138 au<strong>to</strong>psies were performed <strong>and</strong> all the brains were examinedmacro- <strong>and</strong> microscopically. According <strong>to</strong> the results, 29 (21%) suffered from


328 MYCOSEScerebral <strong>to</strong>xoplasmosis; 17 (12%), from cryp<strong>to</strong>coccosis; 2 (1%), from tuberculosis;<strong>and</strong> 1 (0.7%), from c<strong>and</strong>idiasis. In addition, there were cadavers with vascularlesions <strong>and</strong> gliosis; 5% had encephalopathy due <strong>to</strong> HIV (Wainstein et al., 1992).Cryp<strong>to</strong>coccosis often appears in patients weakened by other <strong>diseases</strong> (reticuloendothelialsystem disorders, particularly Hodgkin’s disease) <strong>and</strong> by corticosteroidtreatment. The incubation period is unknown. Pulmonary lesions may precede cerebrallesions by months or years. It is estimated that some 100 deaths per year in theUnited States are due <strong>to</strong> cryp<strong>to</strong>coccosis.Intravenous amphotericin B in doses of 0.4–0.6 mg/kg per day for six weeks canbe effective in many cases. Recently, the preferred therapy is a combination of intravenousamphotericin in reduced doses <strong>and</strong> oral flucy<strong>to</strong>sine. This combination is notindicated for AIDS patients due <strong>to</strong> the early development of signs of flucy<strong>to</strong>sine poisoning.Fluconazole is useful for preventing relapses after administering amphotericinB (Diamond, 1990; Benenson, 1990).The Disease in Animals: The disease has been recognized in cattle, horses,sheep, goats, dogs, cats, nonhuman primates, <strong>and</strong> several species of wild animals (inzoos), but not in birds. Various cases have been described in sheep <strong>and</strong> goats withpulmonary disease <strong>and</strong> mastitis. Of four cases described in goats in WesternAustralia, the pulmonary form predominated in two animals; one had accumulatedfluid in the pleural <strong>and</strong> peri<strong>to</strong>neal cavities, atelectatic lungs, <strong>and</strong> dark red plaque inthe trachea from which C. neoformans was isolated; the fourth animal had analopecic lesion on the head from which a yellow exudate seeped, which showedCryp<strong>to</strong>coccus spp. upon microscopic examination (Chapman et al., 1990). The disseminatedform of the disease is the form most <strong>common</strong>ly diagnosed in dogs <strong>and</strong>cats. Of 21 cases in dogs with a clinical his<strong>to</strong>ry, 13 manifested the meningeal form,4 the nasal form, <strong>and</strong> 1 osteoarticular involvement; the remaining animals hadlesions in other organs. Six cases described in Australia all had the meningeal form(Sut<strong>to</strong>n, 1981). The primary diagnosis in cats has been a disorder of the centralnervous system, with granulomas in the eyes <strong>and</strong> nasal passages, as well as the cutaneousform. Of 29 cats with cryp<strong>to</strong>coccosis, 24 (83%) had the nasal form <strong>and</strong> 15 hadthe cutaneous <strong>and</strong> subcutaneous form. One cat with a significant involvement of thenasal cavity developed meningoencephalitis <strong>and</strong> optical neuritis. Antibodies <strong>to</strong>feline immunodeficiency virus were detected in eight cats. These animals sufferedfrom advanced or disseminated cryp<strong>to</strong>coccosis. C. neoformans var. neoformans wasisolated from 21 cats <strong>and</strong> C. neoformans var. gattii was isolated from 6 cats.Treatment with oral fluconazole yielded very good results. All the cats were curedexcept for one that died four days after treatment began (Malik et al., 1992). Nasal<strong>and</strong> pulmonary tumors with a myxoma<strong>to</strong>us consistency have been observed in variousanimal species. Several outbreaks of mastitis have been confirmed in cows, withvisible abnormalities in the udder <strong>and</strong> changes in the milk. A few cases of meningoencephalitic<strong>and</strong> pulmonary cryp<strong>to</strong>coccosis, cases affecting the frontal sinuses<strong>and</strong> para-orbital area, <strong>and</strong> abortions have been described in horses.Source of Infection <strong>and</strong> Mode of Transmission: Serotypes A <strong>and</strong> D (C. neoformansvar. neoformans) are ubiqui<strong>to</strong>us <strong>and</strong> have been isolated from various environmentalsources, such as soil, certain plants, bird feces, raw milk, <strong>and</strong> fruit juices. Thecausal agent is found frequently in pigeon roosts <strong>and</strong> in soil contaminated by pigeonfeces. The creatinine in pigeon fecal matter serves as a source of nitrogen for C. neo-


CRYPTOCOCCOSIS 329formans,favoring its development <strong>and</strong> prolonging its survival in the soil. Pigeons donot become ill with cryp<strong>to</strong>coccosis.The environmental source of C. neoformans var. gattii was unknown until a fewyears ago. A study conducted in Australia succeeded in isolating var. gattii from 35samples of bark <strong>and</strong> plant remains accumulated under the foliage of a species ofeucalyptus, Eucalyptus camaldulensis. Attempts <strong>to</strong> isolate samples from other eucalyptusspecies were unsuccessful. E. camaldulensis has been exported <strong>to</strong> variouscountries in the Americas, Africa, <strong>and</strong> Asia. The air sample taken from beneath thefoliage demonstrated that the presence of the agent in the air coincided with the eucalyptus’blooming season in late spring. These findings would explain the high incidenceof C. neoformans var. gattii among the aborigines in Australia’s NorthernTerri<strong>to</strong>ry, where these trees are abundant <strong>and</strong> the indigenous population lives in closecontact with them (Ellis <strong>and</strong> Pfeiffer, 1990). Man <strong>and</strong> animals become infected byinhaling dust containing the causal agent. C. neoformans, which has no capsule innature, becomes encapsulated in the lungs, allowing it <strong>to</strong> resist phagocy<strong>to</strong>sis.Although all researchers agree that the infection is contracted through inhalation,there is still debate regarding the infecting element. Some believe it is the yeast formof the agent while others believe it is the basidiospores of the agent’s sexual phase. Ithas also been pointed out that the yeast form would be <strong>to</strong>o large (4 <strong>to</strong> 7 microns) <strong>to</strong>enter the alveoli, while basidiospores measure only about 2 microns (Cohen, 1982).Role of Animals in the Epidemiology of the Disease: There are no known casesof transmission of the disease from animal <strong>to</strong> animal, from animal <strong>to</strong> man, or fromman <strong>to</strong> man, except in the case of a corneal transplant (Beyt <strong>and</strong> Waldman, 1978).Diagnosis: Diagnosis can be made through microscopic observation of encapsulatedC. neoformans in tissues <strong>and</strong> body fluids, <strong>and</strong> can be confirmed by culture. Theuse of culture media <strong>to</strong> differentiate serotypes A <strong>and</strong> D from serotypes B <strong>and</strong> C nowfacilitates serotyping (Salkind <strong>and</strong> Hurd, 1982; Kwon-Chung et al., 1982). Thedirect immunofluorescence test can be used for the same purpose for cultures <strong>and</strong>for some his<strong>to</strong>logical preparations (Kaplan et al., 1981).As the etiologic agent multiplies in the human host, the capsular polysaccharideof C. neoformans neutralizes antibodies. Excess antibodies can be detected in blood<strong>and</strong> urine, as well as in cerebrospinal fluid in cases in which the central nervous systemis affected. Cases that come <strong>to</strong> receive medical attention are frequently faradvanced. Consequently, better results are obtained if the medical examination isdirected <strong>to</strong>ward detecting the specific antigen rather than the antibodies. The platelatex agglutination test with particles sensitized by anticryp<strong>to</strong>coccal globulin is used<strong>to</strong> detect the cryp<strong>to</strong>coccal antigen. The enzyme-linked immunosorbent assay(ELISA) test is also available <strong>to</strong> detect the capuslar polysaccharide antigen of theetiologic agent. This test is much more sensitive than latex agglutination <strong>and</strong> permitsearlier diagnosis (Scott et al., 1980). In patients with meningoencephalitis, a sampleof the cerebrospinal fluid is used for direct microscopic examination <strong>and</strong> a cellcount, another examination with India ink <strong>to</strong> detect encapsulated fungus cells, <strong>and</strong>culture in Sabouroud’s dextrose agar with incubation at 30°C <strong>to</strong> 37°C <strong>to</strong> isolate thefungus. The antigen is sought in serum <strong>and</strong> cerebrospinal fluid.In Engl<strong>and</strong>, 828 HIV-positive patients with fever were examined (in the UnitedKingdom, 85% of cases occur in immunodeficient individuals, while in the UnitedStates, 50% of patients apparently have a normal immune system). Sixty-nine of the


330 MYCOSES828 patients had meningitis. The cryp<strong>to</strong>coccal antigen detection test was performedusing the latex technique for the capsulated polysaccharide antigen. The test waspositive in 16 of 17 patients with meningitis <strong>and</strong> with positive cultures (Nelson etal., 1990).A study conducted on 20 cats with cryp<strong>to</strong>coccosis <strong>and</strong> 184 uninfected animalsused the latex agglutination test. The latex particles were sensitized with rabbit antibodies<strong>to</strong> C. neoformans <strong>to</strong> detect the antigen in the cats’ serum. The test was positivein 19 of the 20 cats with cryp<strong>to</strong>coccosis <strong>and</strong> in none of the controls (Medlean etal., 1990). According <strong>to</strong> some authors, the test has prognostic value in humans, inthat a progressive disease is accompanied by a rise in titer, whereas there is generallya decline in the agglutinating titer when there is clinical improvement.Control: There are no specific measures for preventing the disease. It is important<strong>to</strong> control underlying <strong>diseases</strong> <strong>and</strong> <strong>to</strong> reduce prolonged treatment with corticosteroidsas much as possible.Controlling the pigeon population might prevent some cases. Human exposure <strong>to</strong>accumulations of pigeon excrement should be avoided, particularly on windowsills,in roosts, perches, <strong>and</strong> nests. Removal of pigeon excrement should be preceded bychemical decontamination or by wetting down with water or oil <strong>to</strong> preventaerosolization (Benenson, 1990).BibliographyAinsworth, G.C., P.K.C. Austwick. Fungal Diseases of Animals. 2nd ed. Farnham Royal,Slough, United Kingdom: Commonwealth Agricultural Bureau; 1973.Bava, A.J., R. Negroni. Características epidemiológicas de 106 casos de crip<strong>to</strong>cocosis diagnosticadosen la República Argentina entre 1981–1990. Rev Inst Med Trop Sao Paulo34:335–340, 1992.Bava, A.J., R. Negroni, A.M. Robles, et al. Características epidemiológicas de 71 casos decrip<strong>to</strong>cocosis diagnósticos en diferentes centros asistenciales de la ciudad de Buenos Aires ysus alrededores durante 1991. Infect Microbiol Clin 4:85–89, 1992.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Beyt, B.E., Jr., S.R. Waltman. Cryp<strong>to</strong>coccal endophthalmitis after corneal transplantation.N Engl J Med 298:825–826, 1978.Chapman, H.M., W.F. Robinson, J.R. Bol<strong>to</strong>n, J.P. Robertson. Cryp<strong>to</strong>coccus neoformansinfection in goats. Aust Vet J 67:263–265, 1990.Cohen, J. The pathogenesis of cryp<strong>to</strong>coccosis. J Infect 5:109–116, 1982.Diamond, R.D. Cryp<strong>to</strong>coccus neoformans. In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E.Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: ChurchillLivings<strong>to</strong>ne, Inc.; 1990.Drutz, D.J. The mycoses. In: Wyngarden, J.B., L.H. Smith, Jr., eds. Cecil Textbook ofMedicine. 16th ed. Philadelphia: Saunders; 1982.Ellis, D.H. Cryp<strong>to</strong>coccus neoformans var. gattii in Australia. J Clin Microbiol 25:430–431, 1987.Ellis, D.H., T.J. Pfeiffer. Natural habitat of Cryp<strong>to</strong>coccus neoformans var. gattii. J ClinMicrobiol 28:1642–1644, 1990.Fromtling, R.A., S. Shadomy, H.J. Shadomy, W.E. Dismukes. Serotypes B/C Cryp<strong>to</strong>coccusneoformans isolated from patients in nonendemic areas. J Clin Microbiol 16:408–410, 1982.


CRYPTOCOCCOSIS 331Gordon, M.A. Current status of serology for diagnosis <strong>and</strong> prognostic evaluation of opportunisticfungus infections. In: Pan American Health Organization. Proceedings of the ThirdInternational Conference on the Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1975. (ScientificPublication 304).Kaplan, W., S.L. Bragg, S. Crane, D.G. Ahearn. Serotyping Cryp<strong>to</strong>coccus neoformans byimmunofluorescence. J Clin Microbiol 14:313–317, 1981.Kaufman, L., S. Blumer. Cryp<strong>to</strong>coccosis: The awakening giant. In: Pan American HealthOrganization. Proceedings of the Fourth International Conference on Mycoses: The Black <strong>and</strong>White Yeasts. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1978. (Scientific Publication 356).Kwon-Chung, K.J., J.E. Bennett. Epidemiologic differences between the two varieties ofCryp<strong>to</strong>coccus neoformans. Am J Epidemiol 120:123–130, 1984.Kwon-Chung, K.J., I. Polacheck, J.E. Bennett. Improved diagnostic medium for separationof Cryp<strong>to</strong>coccus neoformans var. neoformans (serotypes A <strong>and</strong> D) <strong>and</strong> Cryp<strong>to</strong>coccus neoformansvar. gattii (serotypes B <strong>and</strong> C). J Clin Microbiol 15:535–537, 1982.Liss, H.P., D. Riml<strong>and</strong>. Asymp<strong>to</strong>matic cryp<strong>to</strong>coccal meningitis. Am Rev Resp Dis124:88–89, 1981.Malik, R., D.I. Wigney, D.B. Muir, et al. Cryp<strong>to</strong>coccosis in cats: Clinical <strong>and</strong> mycologicalassessment of 29 cases <strong>and</strong> evaluation of treatment using orally administered flucanozole. JMed Vet Mycol 30:133–144, 1992.Medlean, L., M.A. Marks, J. Brown, W.L. Borges. Clinical evaluation of a cryp<strong>to</strong>coccalantigen latex agglutination test for diagnosis of cryp<strong>to</strong>coccosis in cats. J Am Vet Med Assoc196:1470–1473, 1990.Muchmore, H.G., F.G. Fel<strong>to</strong>n, S.B. Salvin, E.R. Rhoades. Ecology <strong>and</strong> epidemiology ofcryp<strong>to</strong>coccosis. In: Pan American Health Organization. Proceedings: InternationalSymposium on Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1970. (Scientific Publication 205).Negroni, P. Micosis cutáneas y viscerales. 5.ª ed. Buenos Aires: López; 1972.Nelson, M.R., M. Bower, D. Smith, et al. The value of serum cryp<strong>to</strong>coccal antigen in thediagnosis of cryp<strong>to</strong>coccal infection in patients infected with the human immunodeficiencyvirus. J Infect 21:175–181, 1990.Pathmanathan, R., T.S. Soo-Hoo. Cryp<strong>to</strong>coccosis in the University Hospital Kuala Lumpur<strong>and</strong> review of published cases. Trans Roy Soc Trop Med Hyg 76:21–24, 1982.Salkind, I.F., N.J. Hurd. New medium for differentiation of Cryp<strong>to</strong>coccus neoformansserotype pairs. J Clin Microbiol 15:169–171, 1982.Scott, E.N., H.G. Muchmore, F.G. Fel<strong>to</strong>n. Comparison of enzyme immunoassay <strong>and</strong> latexagglutination methods for detection of Cryp<strong>to</strong>coccus neoformans antigen. Am J Clin Pathol73:790–794, 1980.Sut<strong>to</strong>n, R.H. Cryp<strong>to</strong>coccosis in dogs: A report on 6 cases. Aust Vet J 57:558–564, 1981.Wainstein, M.V., L. Ferreira, L. Wolfenbuttel, et al. Achados neuropa<strong>to</strong>lógicos na síndromeda imunodeficiência adquirida (SIDA): revisão de 138 casos. Rev Soc Brasil Med Trop25:95–99, 1992.


332 MYCOSESDERMATOPHYTOSISICD-10 B35Synonyms: Tinea, derma<strong>to</strong>mycosis, ringworm.Etiology: Several species of Microsporum <strong>and</strong> Trichophy<strong>to</strong>n <strong>and</strong> the speciesEpidermophy<strong>to</strong>n floccosum. Ecologically <strong>and</strong> epidemiologically, three groups ofspecies are distinguished according <strong>to</strong> the reservoir: anthropophilic, zoophilic, <strong>and</strong>geophilic. This discussion will consider only zoophilic species transmissible <strong>to</strong> man.Derma<strong>to</strong>phytes were formerly considered imperfect fungi, Fungi imperfecti orDeuteromycotyna. However, several species have been shown <strong>to</strong> reproduce sexually.The most important zoophilic species are Microsporum canis (whose perfect statereceived the name Nannizzia otae), Trichophy<strong>to</strong>n mentagrophytes (Arthrodermabenhamiae), <strong>and</strong> T. verrucosum. Species of more limited interest are M. equinum, T.equinum, M. gallinae, M. nanum, M. persicolor, <strong>and</strong> T. simii. The species T. mentagrophytesis subdivided in<strong>to</strong> two varieties: T. mentagrophytes var. erinacei <strong>and</strong> var.quinckeanum.The infecting element is the arthrospore (an asexual spore formed in the hyphae<strong>and</strong> released when these break down) of the parasitic phases. Conidia that form inorganic material substrates (where the fungus may form sexual <strong>and</strong> asexual spores)may also be infective.A notable characteristic is that the hyphae <strong>and</strong> spores are highly resistant indesquamated epithelium, where they may remain viable for several months or evenyears if they don’t dry up.Geographic Distribution: Among the zoophilic species, M. canis, T. verrucosum,T. equinum, <strong>and</strong> T. mentagrophytes are distributed worldwide. T. mentagrophytesvar. erinacei has limited distribution (France, Great Britain, Italy, <strong>and</strong> NewZeal<strong>and</strong>) <strong>and</strong> T. simii is limited <strong>to</strong> Asia. The geographic distribution of these fungidepends on the dispersion of the host animals. The host hedgehog of T. mentagrophytesvar. erinacei exists only in Europe <strong>and</strong> in New Zeal<strong>and</strong>, where it was introducedfrom Europe. The abundance or rarity of a derma<strong>to</strong>phyte species dependslargely on the rural or urban habitat <strong>and</strong> the relationship between man <strong>and</strong> animals.M. canis is a fungus that occurs primarily in urban areas where its natural hosts, thedog <strong>and</strong> cat, are abundant <strong>and</strong> in close contact with humans. In contrast, T. verrucosumis found in rural areas, particularly among stabled cattle, i.e., generally in areaswith cold or temperate climates.Occurrence in Man: Derma<strong>to</strong>phytic infections are <strong>common</strong>, but their exact prevalenceis unknown. The disease is not notifiable <strong>and</strong>, moreover, many people withminor infections do not see a doc<strong>to</strong>r. Most of the data come from derma<strong>to</strong>logists,mycology labora<strong>to</strong>ries, <strong>and</strong> epidemiologic investigations. Economically advancedcountries have experienced a marked reduction in some species of anthropophilic derma<strong>to</strong>phytes.This is true of M. audouinii, which causes epidemic outbreaks of tineacapitis. In such countries, zoophilic derma<strong>to</strong>phytes are now much more significant. Astudy conducted in Engl<strong>and</strong> on 23 families <strong>to</strong> evaluate the prevalence of the infectionamong family members who were in contact with clinically or sub-clinically infectedyoung cats found that 46 (50%) out of 92 individuals contracted the infection due <strong>to</strong>


DERMATOPHYTOSIS 333M. canis. The percentage of adults infected was 44.2% <strong>and</strong> that of children <strong>and</strong> youngpeople was 80% (12 out of 15) (Pepin <strong>and</strong> Oxenham, 1986). A retrospective study of1,717 Ministry of Agriculture veterinarians in the United Kingdom found that derma<strong>to</strong>phy<strong>to</strong>siswas the most <strong>common</strong> zoonosis, with a prevalence of 24% (Constable<strong>and</strong> Hering<strong>to</strong>n, cited by Pepin <strong>and</strong> Oxenham, 1986). In northeast Madrid (Spain), theannual incidence of derma<strong>to</strong>phy<strong>to</strong>sis was found <strong>to</strong> be 84 cases per 10,000 inhabitants,<strong>and</strong> the most frequent agents in 135 patients were Epidermophy<strong>to</strong>n floccosum, ananthropophilic derma<strong>to</strong>phyte (35.5%), Microsporum canis (26.6%), <strong>and</strong>Trichophy<strong>to</strong>n mentagrophytes (20.7%) (Cuadros et al., 1990). A retrospective studyconducted in Argentina on 1,225 samples of superficial mycoses (95% adult patients<strong>and</strong> 5% children) indicated that 60% were derma<strong>to</strong>phytes. The most <strong>common</strong> agentwas T. rubrum (66.6%), followed by T. mentagrophytes (20%), M. canis (8%), <strong>and</strong>others (Canteros et al., 1993). In Peru, it is likely that zoophilic species are responsiblefor 21% of human derma<strong>to</strong>mycoses (Gómez P<strong>and</strong>o <strong>and</strong> Ma<strong>to</strong>s Díaz, 1982). Astudy conducted in India found that T. verrucosum <strong>and</strong> T. mentagrophytes var. mentagrophyteswere responsible for 56 (38.6%) of 145 human isolates <strong>and</strong> for 50(53.8%) of the 93 human cases in the rural area (Chatterjee et al., 1980).Many human cases originated in Hungary, in a nursery of 5,500 rabbits. Over thecourse of six months, all the rabbits were infected by T. mentagrophytes var. mentagrophytes(var. granulosum) <strong>and</strong> 38 human cases appeared among workers <strong>and</strong> theirfamilies (Szili <strong>and</strong> Kohalmi, 1983).Occurrence in Animals: In recent years, epidemiologic studies have demonstratedthat derma<strong>to</strong>phytic infection in animals is very <strong>common</strong>. Tinea occurs morefrequently among stabled animals than those kept in open pastures throughout theyear.Infection by M. canis is very <strong>common</strong> in cats <strong>and</strong> dogs <strong>and</strong> is often asymp<strong>to</strong>matic.In Lima (Peru) <strong>and</strong> its environs, M. canis was found in 12 (15%) of 79 cats withoutapparent lesions examined <strong>and</strong> T. mentagrophytes in 8 (10%). M. canis was isolatedfrom 17 (3.9%) of 432 samples from dogs, <strong>and</strong> T. mentagrophytes from 22 (5%)(Gómez P<strong>and</strong>o <strong>and</strong> Ma<strong>to</strong>s Díaz, 1982). In the United Kingdom, in 1,368 derma<strong>to</strong>phytesisolated between 1956 <strong>and</strong> 1991, Microsporum canis was diagnosed in 92%of infected cats <strong>and</strong> in 65% of dogs (Sparkes et al., 1993). Long-haired cats <strong>and</strong> dogsunder one year of age were most affected (Sparkes et al., 1993).The cat is the most <strong>common</strong> host <strong>and</strong> reservoir of M. canis. Some studies havefound infection in between 6.5% <strong>and</strong> 88% or more of the cats examined. These studieswere conducted in areas where the cats were in contact with other cats. A completelydifferent picture was obtained in a study conducted at the University ofWisconsin (USA). Fifteen genera of fungi, 13 of which were considered saprophytes,were isolated from the skin of 172 cats that lived alone with their owners. T.rubrum, considered an anthropophilic derma<strong>to</strong>phyte, was isolated from 14 cats; T.gypseum <strong>and</strong> M. vanbreuseghemii, both geophilic, were isolated from 1 cat each; butM. canis was isolated from none. T. rubrum is a <strong>common</strong> agent in human derma<strong>to</strong>phy<strong>to</strong>sis,but the role that cats might play in its transmission <strong>to</strong> man is unknown(Moriello <strong>and</strong> De Boer, 1991).The Disease in Man: Derma<strong>to</strong>phy<strong>to</strong>sis, or tinea, is a superficial infection of thekeratinized parts of the body (skin, hair, <strong>and</strong> nails). As a general rule, zoophilic <strong>and</strong>geophilic derma<strong>to</strong>phytes produce more acute inflamma<strong>to</strong>ry lesions than the anthro-


334 MYCOSESpophilic species, which are parasites better adapted <strong>to</strong> man. The Microsporumspecies cause most cases of tinea capitis <strong>and</strong> tinea corporis, but are rarely responsiblefor infection of the nails (onychomycosis) or skin folds (intertrigo). However, theTrichophy<strong>to</strong>n species can affect the skin in any part of the body.There are two varieties of T. mentagrophytes: an anthropophilic variety (var. interdigitale)that is relatively nonvirulent in humans <strong>and</strong> localizes in the feet (athlete’sfoot), <strong>and</strong> a zoophilic variety (morphologically granular) that causes a very inflamma<strong>to</strong>ryderma<strong>to</strong>phy<strong>to</strong>sis on different areas of the human body. The zoophilic varietyis usually found in rodents, cats, dogs, <strong>and</strong> other animals. Transmission <strong>to</strong> man isprobably caused by contamination of his habitat by hair from infected animals.Several epidemic outbreaks of inflamma<strong>to</strong>ry derma<strong>to</strong>phy<strong>to</strong>ses on different parts ofthe body among the U.S. troops in Vietnam were caused by T. mentagrophytes var.mentagrophytes (var. granulosum). About one-fourth of the rats trapped in the vicinityof military camps were infected with strains of the same variety of fungus. Amongthe inhabitants of the region, the disease was seen only in children, suggesting thatadults were probably immunized by infections contracted during childhood.Currently, M. canis is one of the principal etiologic agents of tinea <strong>and</strong>, in manycountries, has displaced the anthropophilic species M. audouinii as the cause oftinea capitis. In South America, M. canis is the most <strong>common</strong> of the microspora.The incubation period of the disease is one <strong>to</strong> two weeks. Tinea of the scalp ismost frequent among those aged 4 <strong>to</strong> 11 years <strong>and</strong> its incidence is higher amongmales. The disease begins with a small papule, the hair becomes brittle, <strong>and</strong> theinfection spreads peripherally, leaving scaly, bald patches. Suppurative lesions (kerions)are frequent when the fungus is of animal origin. Tinea caused by M. canisheals spontaneously during puberty.Suppurative tinea barbae, which affects rural populations, is caused by T. mentagrophytesof animal origin. However, in the United States dry tinea barbae is causedby T. mentagrophytes of human origin <strong>and</strong> by T. rubrum (Silva-Hunter et al., 1981).Tinea corporis is characterized by flat lesions that tend <strong>to</strong> be annular. The bordersare reddish <strong>and</strong> may be raised, with microvesicles or scales.Tinea corporis in children is usually an extension of tinea capitis <strong>to</strong> the face <strong>and</strong>is caused by M. canis or M. audouinii. Active lesions may also appear on the wrists<strong>and</strong> neck of mothers or young adults who have contact with the infected child. Tineacorporis in adults, occurring primarily on the limbs <strong>and</strong> <strong>to</strong>rso, is chronic in nature<strong>and</strong> usually is caused by the anthropophilic derma<strong>to</strong>phyte T. rubrum (Silva-Hunteret al., 1981).Tinea pedis (athlete’s foot), the incidence of which is increasing worldwide, iscaused by anthropophilic species of Trychophy<strong>to</strong>n <strong>and</strong>, <strong>to</strong> a lesser extent, byEpidermophy<strong>to</strong>n floccosum (also anthropophilic).In AIDS patients, mycosis caused by T. mentagrophytes <strong>and</strong> M. canis can be cutaneous<strong>and</strong> disseminated (Lowinger-Seoane et al., 1992). AIDS patients may sufferfrom extensive derma<strong>to</strong>phy<strong>to</strong>sis caused by a fungus as rare in humans as M. gallinae,a zoophilic derma<strong>to</strong>phyte; there are only seven known cases, all of them localized(del Palacio et al., 1992).The recommended treatment is <strong>to</strong>pical application of antimycotics. The azoles(miconazole, clotrimazole, econazole, bifonazole, oxiconazole, tioconazaole, <strong>and</strong>others) are used most frequently. These antimycotics produce good results in allforms of dermal tinea caused by zoophilic derma<strong>to</strong>phytes.


DERMATOPHYTOSIS 335Topical treatment should continue for two <strong>to</strong> four weeks. Naftifine is anotherpowerful antimycotic (Hay, 1990).The Disease in Animals: The most important species considered reservoirs ofderma<strong>to</strong>phytes transmissible <strong>to</strong> humans are cats, dogs, cattle, horses, <strong>and</strong> rodents.CATS AND DOGS: The most important etiologic agent in these animals is M. canis.This derma<strong>to</strong>phyte species is very well adapted <strong>to</strong> cats <strong>and</strong> approximately 90% ofinfected animals manifest no apparent lesions. When lesions do occur, they appearprimarily on the face <strong>and</strong> paws.Lesions are frequent <strong>and</strong> apparent in dogs <strong>and</strong> may appear on any part of the bodyin the form of tinea circinata (ringworm).Dogs <strong>and</strong> cats may also be infected by other derma<strong>to</strong>phytes, particularly T.mentagrophytes.CATTLE: The principal etiologic agent of tinea in cattle is T. verrucosum (T. faviforme,T. ochraceum, T. album, <strong>and</strong> T. discoides). The disease is more <strong>common</strong> incountries where animals are kept in stables during winter, <strong>and</strong> its incidence is higherin calves than in adults. Lesions may be as small as 1 cm in diameter or may coverextensive areas; they are most frequently located on the face <strong>and</strong> neck, but lesions arealso found with some frequency on other parts of the body, such as the flanks <strong>and</strong> legs.The lesion is initially characterized by grayish white, dry areas with a few brittle hairs.The lesion then thickens <strong>and</strong> resembles a light brown scab. The scab falls off, leavingan alopecic area. The condition clears up spontaneously within two <strong>to</strong> four months.HORSES: Derma<strong>to</strong>phy<strong>to</strong>sis in horses is caused by T. equinum <strong>and</strong> M. equinum; thelatter is rare in the Americas. Lesions are usually found in areas where the harnesscauses friction. They are dry, bald, covered with scales, <strong>and</strong> the skin is thickened.Colts are the most susceptible. Infections caused by Trichophy<strong>to</strong>n equinum are usuallymore severe, with pruritus <strong>and</strong> exudative lesions causing the hair <strong>to</strong> stick<strong>to</strong>gether in clumps. When they drop off, they leave alopecic areas. Infections due <strong>to</strong>M. equinum cause less serious lesions with small scaly areas with brittle hairs.RODENTS AND LAGOMORPHS: Tinea favus of mice, caused by T. mentagrophytesvar. quinckeanum, is widely distributed throughout the world <strong>and</strong> is transmissible <strong>to</strong>domestic animals <strong>and</strong> man. The lesion is white <strong>and</strong> scabby <strong>and</strong> localized on the head<strong>and</strong> trunk. T. mentagrophytes (var. mentagrophytes) is another derma<strong>to</strong>phyte <strong>common</strong><strong>to</strong> rodents. Labora<strong>to</strong>ry mice <strong>and</strong> guinea pigs are mostly infected by T. mentagrophytes,<strong>and</strong> may not have apparent lesions; the agent’s presence is often detectedwhen humans contract the infection. It is also transmissible <strong>to</strong> dogs.Derma<strong>to</strong>phy<strong>to</strong>sis in rabbits is also caused by T. mentagrophytes <strong>and</strong> usuallyoccurs in animals that have recently been weaned. Scabby areas of alopecia are seenclinically around the eyes <strong>and</strong> nose. Secondary lesions appear on the feet. This diseaseis self-limiting.SHEEP AND GOATS: Tinea is rare in these species. The lesions localize on the head<strong>and</strong> face. The most frequent agent is T. verrucosum. The lesions are limited <strong>to</strong> areasof the head covered by hair; they are circular, balding, <strong>and</strong> have thick scabs. Twooutbreaks of derma<strong>to</strong>phy<strong>to</strong>sis caused by M. canis were described in Australia. In thefirst outbreak, transmission was attributed <strong>to</strong> cats <strong>and</strong> <strong>to</strong> the use of contaminatedshearing implements. In the second, with 20% of 90 sheep infected, it was not pos-


336 MYCOSESsible <strong>to</strong> determine how the infection had been introduced, but its spread throughoutthe establishment was undoubtedly due <strong>to</strong> shearing implements <strong>and</strong> close contactamong the animals immediately after being sheared (Jackson et al., 1991).SWINE: The most <strong>common</strong> agent of swine tinea is M. nanum. Infection has beenconfirmed in Australia, Canada, Cuba, Kenya, Mexico, New Guinea, New Zeal<strong>and</strong>,<strong>and</strong> the United States. This derma<strong>to</strong>phyte was isolated in only a few human cases.The lesion is characterized by a wrinkled area covered by a thin, brown scab thatdetaches easily. M. nanum lives as a soil saprophyte in areas where swine are raised<strong>and</strong> is classified as geophilic.FOWL: Tinea favus in hens occurs sporadically throughout the world <strong>and</strong> is rarelytransmitted <strong>to</strong> man. Its agent is T. gallinae.Source of Infection <strong>and</strong> Mode of Transmission: The natural reservoirs ofzoophilic derma<strong>to</strong>phytes are animals. Transmission <strong>to</strong> man occurs through contactwith an infected animal (either sick or a carrier) or indirectly through spores containedin the hair <strong>and</strong> dermal scales shed by the animal. Derma<strong>to</strong>phytes remain viablein shed epithelium for several months or even years. The same animal can infect severalpeople within a family, but a zoophilic derma<strong>to</strong>phyte does not usually spreadfrom person <strong>to</strong> person <strong>and</strong>, unlike the anthropophilic derma<strong>to</strong>phytes, does not causeepidemic tinea. Cases of human-<strong>to</strong>-human transmission of M. canis have beenobserved, but the agent loses its infectiveness for man after a few intermediaries(Padhye, 1980). A nosocomial infection was described in a nursery for newborns.Although tinea of the scalp is <strong>common</strong> among children, it is rarely found in newborns.The <strong>common</strong> source of the infection turned out <strong>to</strong> be a nurse who had an indolentinfection due <strong>to</strong> M. canis (Snider et al., 1993). T. verrucosum, whose principalreservoir is cattle, is found in infections in rural populations. A study conducted inSwitzerl<strong>and</strong> found that 14% of those working with infected cattle contracted derma<strong>to</strong>phy<strong>to</strong>siscaused by T. verrucosum (Haub, as reported <strong>to</strong> Gudding et al., 1991).This mycosis also has economic consequences, in that skins from the infected animaldepreciate in value. It is a reportable disease in Norway. In contrast, M. canis is transmittedby cats <strong>and</strong> dogs <strong>to</strong> urban <strong>and</strong> rural populations. The cat is considered the principalsource of infection for humans due <strong>to</strong> the cus<strong>to</strong>m of picking up <strong>and</strong> petting acat, as well as <strong>to</strong> its high rate of infection. Cats can also host the anthropophilic derma<strong>to</strong>phyte,T. rubrum, in their hair, but it has not been demonstrated that they cantransmit it <strong>to</strong> man. Infection due <strong>to</strong> T. mentagrophytes var. mentagrophytes (var. granulosum)<strong>and</strong> T. mentagrophytes var. quinckeanum is indirectly transmitted fromrodents <strong>to</strong> man via residues of shed epithelium in the environment. Cats <strong>and</strong> dogs canalso become infected by these derma<strong>to</strong>phytes in the same way or by direct contactwhen they hunt rodents <strong>and</strong> can, in turn, transmit the infection <strong>to</strong> man.Animal-<strong>to</strong>-animal transmission occurs in the same ways. Crowding <strong>and</strong> reducedorganic resistance influence the incidence of infection.Role of Animals in the Epidemiology of the Disease: Animals are the reservoirof zoophilic derma<strong>to</strong>phytes <strong>and</strong> the source of infection for man. As in other<strong>zoonoses</strong>, human-<strong>to</strong>-human transmission is rare. Transmission of anthropophilicderma<strong>to</strong>phytes from humans <strong>to</strong> animals is also rare.The derma<strong>to</strong>phyte M. gypseum is the causal agent of sporadic cases of tinea inhumans <strong>and</strong> animals; its reservoir is the soil (geophilic).


DERMATOPHYTOSIS 337Diagnosis: Clinical diagnosis can be confirmed by the following methods: a)microscopic observation of hair <strong>and</strong> scales from lesions; this method can provide adiagnosis at the genus level, since the spores surround the hair shaft in an irregularmosaic when infection is due <strong>to</strong> Microsporum <strong>and</strong> are arranged in chains wheninfection is due <strong>to</strong> Trichophy<strong>to</strong>n; b) the use of Wood’s light (filtered ultravioletlight), under which hair infected by many species of Microsporum exhibits a brightblue-green fluorescence; c) isolation in culture media, the only method that permitsidentification of the species.Control: Prevention of human derma<strong>to</strong>phy<strong>to</strong>ses caused by zoophilic speciesshould be based on controlling the infection in animals, although this is difficult <strong>to</strong>accomplish. Avoiding contact with animals that are obviously sick can prevent a certainpercentage of human cases. These animals should be isolated <strong>and</strong> treated with<strong>to</strong>pical antimycotics or griseofulvin administered orally. Remains of hair <strong>and</strong> scalesshould be burned <strong>and</strong> rooms, stables, <strong>and</strong> all utensils should be disinfected.Apparently healthy cats can be examined with Wood’s light. Controlling the rodentpopulation is a useful measure.In cold climates where animals are stabled over long periods of time, derma<strong>to</strong>phy<strong>to</strong>sescan be a problem in cattle <strong>and</strong> horses. Man <strong>and</strong> animals respond <strong>to</strong> infectionwith a humoral <strong>and</strong> cellular immunity, as has been demonstrated by experimentsas well as by the observation that animals once infected are protected againstreinfection. Two vaccines were developed in the former Soviet Union: one for cattle,made from an attenuated strain of T. verrucosum, <strong>and</strong> another for horses, madefrom T. equinum. Both vaccines yielded satisfac<strong>to</strong>ry results in preventing derma<strong>to</strong>phy<strong>to</strong>ses.The vaccine was used in Norway in 200,000 cattle with very good results(Aamodt et al., 1982). An eradication program was established in Gausdal, Norway;vaccination was required for all cattle for a period of six years, followed by voluntaryvaccination thereafter. The prevalence of infected herds was 70% <strong>and</strong> eradicationwas achieved in 1987. A live attenuated vaccine was used (two doses with aninterval of 14 days) along with disinfection of stables, isolation of infected animals,<strong>and</strong> other hygiene methods (Gudding et al., 1991).BibliographyAamodt, O., B. Naess, O. S<strong>and</strong>vik. Vaccination of Norwegian cattle against ringworm. ZblVet Med B 29:451–456, 1982.Ainsworth, G.C., P.K.C. Austwick. Fungal Diseases of Animals. 2nd ed. Farnham Royal,Slough, United Kingdom: Commonwealth Agricultural Bureau; 1973.Allen, A.M., D. Taplin. Epidemiology of cutaneous mycoses in the tropics <strong>and</strong> subtropics:Newer concepts. In: Pan American Health Organization. Proceedings of the ThirdInternational Conference on the Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1975. (ScientificPublication 304).Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Canteros, C.E., G.O. Davel, W. Vivot, S. D’Amico. Incidencia de los distin<strong>to</strong>s agentes etiológicosde micosis superficiales. Rev Argent Microbiol 25:129–135, 1993.


338 MYCOSESChatterjee, A., D. Chat<strong>to</strong>padhyay, D. Bhattacharya, A.K. Dutta, D.N. Sen Gupta. Some epidemiologicalaspects of zoophilic derma<strong>to</strong>phy<strong>to</strong>sis. Int J Zoonoses 7:19–33, 1980.Chmel, L. Epidemiological aspects of zoophilic derma<strong>to</strong>phytes. In: Chmel, L., ed. RecentAdvances in Human <strong>and</strong> Animal Mycology. Bratislava: Slovak Academy of Sciences; 1967.Cuadros, J.A., J. García, J.I. Alos, R. González-Palacios. Derma<strong>to</strong>fi<strong>to</strong>sis en un mediourbano: un estudio prospectivo de 135 casos. Enferm Infecc Microbiol Clin 8:429–433, 1990.del Palacio, A., M. Pereiro-Miguens, C. Gimeno, et al. Widespread derma<strong>to</strong>phy<strong>to</strong>sis due <strong>to</strong>Microsporum (Trichophy<strong>to</strong>n) gallinae in a patient with AIDS: A case report from pain. ClinExp Derma<strong>to</strong>l 17:449–453, 1992.Emmons, C.W. Mycoses of animals. Adv Vet Sci 2:47–63, 1955.English, M.P. The epidemiology of animal ringworm in man. Br J Derma<strong>to</strong>l86(Suppl)8:78–87, 1972.Gentles, J.C. Ringworm. In: Graham-Jones, O., ed. Some Diseases of AnimalsCommunicable <strong>to</strong> Man in Britain. Oxford: Pergamon Press; 1968.Georg, L.K. Animal Ringworm in Public Health, Diagnosis <strong>and</strong> Nature. Atlanta, Georgia:U.S. Centers for Disease Control <strong>and</strong> Prevention; 1960. (Public Health ServicePublication 727).Gómez P<strong>and</strong>o, V., J. Ma<strong>to</strong>s Díaz. Derma<strong>to</strong>fi<strong>to</strong>s: aspec<strong>to</strong>s epidemiológicos. Bol Inf Col MedVet Peru 17:16–19, 1982.Gudding, R., B. Naess, O. Aamodt. Immunisation against ringworm in cattle. Vet Rec128:84–85, 1991.Hay, R.J. Derma<strong>to</strong>phy<strong>to</strong>sis <strong>and</strong> other superficial mycoses. In: M<strong>and</strong>ell, G.L., R.G. Douglas,Jr., J.E. Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York:Churchill Livings<strong>to</strong>ne Inc.; 1990.Jackson, R.B., B.F. Peel, C. Donaldson-Wood. Endemic Microsporum canis infection in asheep flock. Aust Vet J 68:122, 1991.Lowinger-Seoane, M., J.M. Torres-Rodríguez, N. Madrenys-Brunet, et al. Extensive derma<strong>to</strong>phy<strong>to</strong>siscaused by Trichophy<strong>to</strong>n mentagrophytes <strong>and</strong> Microsporum canis in a patientwith AIDS. Mycopathologia 120:143–146, 1992.Moriello, K.A., D.J. De Boer. Fungal flora of the coat of pet cats. Am J Vet Res 52:602–606, 1991.Negroni, P. Micosis cutáneas y viscerales. 5.ª ed. Buenos Aires: López; 1972.Padhye, A.A. Cutaneous mycoses. In: S<strong>to</strong>enner, J., W. Kaplan, M. Torten, eds. Section A,Vol 2: CRC H<strong>and</strong>book Series in Zoonoses. Boca Ra<strong>to</strong>n: CRC Press; 1980.Pepin, G.C., P.K.C. Austwick. Skin <strong>diseases</strong> of domestic animals. II. Skin disease, mycologicalorigin. Vet Rec 82:208–214, 1968.Pepin, G.A., M. Oxenham. Zoonotic derma<strong>to</strong>phy<strong>to</strong>sis (ringworm) [letter]. Vet Rec118:110–111, 1986.Raubitscheck, F. Fungal <strong>diseases</strong>. In: Van der Hoeden, J., ed. Zoonoses. Amsterdam:Elsevier; 1964.Rebell, G., D. Taplin. Derma<strong>to</strong>phytes: Their Recognition <strong>and</strong> Identification. Miami:University of Miami Press; 1970.Sarkisov, A.K. New methods of control of derma<strong>to</strong>mycoses <strong>common</strong> <strong>to</strong> animals <strong>and</strong> man.In:Lysenko, A., ed. Vol 2: Zoonoses Control. Moscow: Centre of International Projects; 1982.Silva-Hunter, M., I. Weitzman, S.A. Rosenthal. Cutaneous mycoses (derma<strong>to</strong>mycoses). In:Balows, A., W.J. Hausler, Jr., eds. Diagnostic Procedures for Bacterial, Mycotic <strong>and</strong> ParasiticInfections. 6th ed. Washing<strong>to</strong>n, D.C.: American Public Health Association; 1981.Smith, J.M.B. Superficial <strong>and</strong> cutaneous mycoses. In: Hubbert, W.T., W.F. McCulloch, P.R.Schnurrenberger, eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield:Thomas; 1975.Snider, R., S. L<strong>and</strong>ers, M.L. Levy. The ringworm riddle: An outbreak of Microsporum canisin the nursery. Pediatr Infect Dis J 12:145–148, 1993.


HISTOPLASMOSIS 339Sparkes, A.H., T.J. Gruffyd-Jones, S.E. Shaw, et al. Epidemiological <strong>and</strong> diagnostic featuresof canine <strong>and</strong> feline derma<strong>to</strong>phy<strong>to</strong>sis in the United Kingdom from 1956 <strong>to</strong> 1991. Vet Rec133:57–61, 1993.Szili, M., I. Kohalmi. Endemic Trichophy<strong>to</strong>n mentagrophytes infection of rabbit origin.Mykosen 24:412–420, 1981. Abst Rev Med Vet 64:65, 1983.HISTOPLASMOSISICD-10 B39.0 acute pulmonary his<strong>to</strong>plasmosis capsulati; B39.1 chronicpulmonary his<strong>to</strong>plasmosis capsulati; B39.3 disseminated his<strong>to</strong>plasmosiscapsulati; B39.5 His<strong>to</strong>plasma duboisiiSynonyms: Reticuloendothelial cy<strong>to</strong>mycosis, cavern disease, Darling’s disease.Etiology: His<strong>to</strong>plasma capsulatum, a dimorphic fungus that has a yeast form inthe parasitic phase <strong>and</strong> develops a filamen<strong>to</strong>us mycelium in the saprophytic phase,producing macroconidia <strong>and</strong> microconidia. The yeast form may also be grown in thelabora<strong>to</strong>ry by culturing the fungus in an enriched medium at 37°C. The perfect (orsexual) state of the fungus is also known <strong>and</strong> has been given the name Emmonsielacapsulata.There are two known varieties of the agent: H. capsulatum var. capsulatum <strong>and</strong>H. capsulatum var. duboisii. They are indistinguishable in the mycelial phase but ininfected tissue the yeast-form cells of var. duboisii are much larger (7–15 microns)than those of var. capsulatum (2–5 microns). The tissue reactions they produce arealso different. In regions in which the two varieties of the fungus coexist, the use ofmonoclonal antibodies in the ELISA or Western blot tests has been suggested fordifferentiating them in the yeast phase (Hamil<strong>to</strong>n et al., 1990).Geographic Distribution: Distribution of var. capsulatum is worldwide <strong>and</strong>more abundant in the Americas than other continents. Au<strong>to</strong>chthonous human casesare rare in Europe <strong>and</strong> Asia. The var. duboisii is known only in Africa between20° S <strong>and</strong> 20° N (there are known cases in Madagascar) (Coulanges, 1989), wherethe other variety is known <strong>to</strong> exist as well. Distribution of the fungus in the soil isnot uniform, as some regions are more contaminated than others <strong>and</strong> microfoci existwhere the agent is highly concentrated. The assumption is that endemic areas wouldbe determined by the number of microfoci. As for the duboisii variety, efforts <strong>to</strong>determine its habitat in the environment have been unsuccessful.Occurrence in Man: Judging from the results of the his<strong>to</strong>plasmin intradermaltest, the rate of infection is very high in endemic areas. It has been estimated that inthe United States, where the infection is concentrated in the Missouri, Ohio, <strong>and</strong>Mississippi river basins, 30 million inhabitants have been infected by His<strong>to</strong>plasma<strong>and</strong> some half million people become infected each year (Selby, 1975). The diseaseappears sporadically or in epidemic outbreaks. Isolated cases frequently elude diagnosis.There was an outbreak in 1980 with 138 cases of acute pulmonary disease


340 MYCOSESamong workers in a lime quarry in northern Michigan, an area not consideredendemic (Waldman et al., 1983). Another outbreak occurred in 1978–1979 at theIndianapolis campus of Indiana University, affecting 435 people. Again in1980–1981, an outbreak in an area close <strong>to</strong> the same university affected 51 people(Schlech et al., 1983). His<strong>to</strong>plasmosis is considered the most <strong>common</strong> systemicmycotic infection in the United States (Loyd et al., 1990). There are also endemicregions in Latin America. Although prevalence varies from region <strong>to</strong> region, it hasbeen claimed that the entire population of Latin America lives within or near areaswhere the infection can be contracted (Borelli, 1970). In Mexico, epidemic outbreaksor isolated cases of the disease have been recorded in all but two states. Therewas a study of 11 outbreaks affecting 75 people in 1979, with mortality at 5.3%, <strong>and</strong>12 outbreaks affecting 68 people in 1980. Most of the cases occurred in people whofor occupational, educational, or recreational reasons had visited caves, ab<strong>and</strong>onedmines, <strong>and</strong> tunnels in which bat droppings had accumulated. More than 2,000 largemines have had <strong>to</strong> be ab<strong>and</strong>oned because of the presence of H. capsulatum due <strong>to</strong>large bat colonies (OPS, 1981). There are also endemic areas in Guatemala, Peru,<strong>and</strong> Venezuela (Ajello <strong>and</strong> Kaplan, 1980). In Cuba, three outbreaks, one of whichaffected 521 people, occurred between 1962 <strong>and</strong> 1963. In 1978 there was an outbreakamong students who visited a cave in the province of Havana; more recently,in a cave in the city of Morón, seven of eight spelunkers contracted the disease(González Menocal et al., 1990).Although the infection is <strong>common</strong>, the clinical disease is much less so.Radiography revealed pulmonary calcifications in a high percentage (about 25%) ofpeople reacting <strong>to</strong> his<strong>to</strong>plasmin. Approximately 90% of those who have a positivereaction <strong>to</strong> the his<strong>to</strong>plasmin hypersensitivity skin test are clinically normal.In Africa, there are some 200 known cases of his<strong>to</strong>plasmosis due <strong>to</strong> the duboisiivariety (Coulanges, 1989).Occurrence in Animals: Many species of domestic <strong>and</strong> wild mammals are susceptible<strong>to</strong> the infection. Surveys using the his<strong>to</strong>plasmin test have shown that infectionis frequent in cattle, sheep, <strong>and</strong> horses in endemic areas. Dogs are the animalspecies in which the infection appears most frequently with clinical symp<strong>to</strong>ms. Of14,000 dogs admitted <strong>to</strong> the University of Ohio clinic (USA) over the course of fouryears, his<strong>to</strong>plasmosis was diagnosed in 62 (0.44%) (Cole et al., 1953).The Disease in Man: When conidia are inhaled, they can lodge in the bronchioles<strong>and</strong> alveoli. After a few days, they germinate <strong>and</strong> produce yeasts that are phagocytizedby macrophages where they proliferate. The macrophages move <strong>to</strong>ward themediastinal lymph nodes <strong>and</strong> the spleen. When immunity develops, themacrophages acquire the ability <strong>to</strong> destroy the phagocytized yeasts, <strong>and</strong> the infiltratesin the nodes <strong>and</strong> other infection sites disappear (Loyd et al., 1990). Mostinfections occur asymp<strong>to</strong>matically. The development of the disease depends on thenumber of conidia inhaled <strong>and</strong> on the individual’s cellular immunity. The incubationperiod lasts from 5 <strong>to</strong> 18 days. There are essentially three clinical forms of the disease:acute pulmonary, chronic cavitary pulmonary, <strong>and</strong> disseminated. The acutepulmonary form is the most frequent <strong>and</strong> resembles influenza with febrile symp<strong>to</strong>msthat may last from one day <strong>to</strong> several weeks. A high percentage of patients alsoexperience cough <strong>and</strong> chest pain. In most patients, chest radiographs show nochanges, but in other cases small infiltrates <strong>and</strong> an increase in the hilar <strong>and</strong>


HISTOPLASMOSIS 341mediastinal nodes can be seen. Erythema nodosum or multiforme, diffuse eruption,<strong>and</strong> arthralgia may be present. This form of the disease often goes unnoticed. In mildcases, recovery occurs without treatment, with or without pulmonary calcification.The chronic form of the disease occurs most frequently in people over the age of 40,with a high prevalence among males, <strong>and</strong> almost always with a preexisting pulmonarydisease (particularly emphysema). Its clinical form resembles pulmonarytuberculosis, with cavitation. The course may vary from months <strong>to</strong> years <strong>and</strong> curemay be spontaneous. The disseminated form of the disease is the most serious <strong>and</strong>is seen primarily in the very young or elderly, where it can take an acute or chroniccourse. The acute course occurs primarily among nursing babies (immature immunity)<strong>and</strong> small children <strong>and</strong> is characterized by different degrees ofhepa<strong>to</strong>splenomegaly, fever, <strong>and</strong> prostration. It is often confused with miliary tuberculosis<strong>and</strong> is highly fatal if the patient is not treated. Leukopenia, thrombocy<strong>to</strong>penia,<strong>and</strong> anemia are frequent. The agent can be isolated from blood <strong>and</strong> bone marrow.Between 1934 <strong>and</strong> 1988, the medical literature recorded only 73 pediatric casesof disseminated his<strong>to</strong>plasmosis (Mir<strong>and</strong>a Novales et al., 1993). The symp<strong>to</strong>ma<strong>to</strong>logyin the chronic disseminated form depends on the localization of the fungus(pneumonia, hepatitis, endocarditis, etc.). There is frequently ulceration of themucosa <strong>and</strong> hepa<strong>to</strong>splenomegaly in these cases. It usually occurs in adults, who maysurvive for many years, but it can be fatal if the patient is not treated.Disseminated his<strong>to</strong>plasmosis occurs in immunodeficient patients, includingpatients with AIDS. It is sometimes the first manifestation of the syndrome <strong>and</strong> insome endemic areas it is the most <strong>common</strong> infection in AIDS (Johnson et al., 1988).The forms of the disease <strong>and</strong> their symp<strong>to</strong>ms are very varied. The most frequentclinical symp<strong>to</strong>ms in 27 patients (23 men <strong>and</strong> 4 women) were fever, weight loss,anemia, cutaneous lesions, micronodules in the lungs, hepa<strong>to</strong>splenomegaly, <strong>and</strong>adenomegaly (Negroni et al., 1992). Some cases follow a fulminant course with respira<strong>to</strong>ryinsufficiency; other cases involve encephalopathy (AIDS dementia), gastrointestinalhis<strong>to</strong>plasmosis with intestinal perforation, <strong>and</strong> cutaneous his<strong>to</strong>plasmosiswith papules on the limbs, face, <strong>and</strong> <strong>to</strong>rso.Fifty radiographs of AIDS patients suffering from disseminated his<strong>to</strong>plasmosisindicated no changes in 27 patients <strong>and</strong> different abnormalities (nodular opacities<strong>and</strong> irregular or linear opacities) in 23 patients. Radiographic results in thesepatients were varied <strong>and</strong> nonspecific (Conces et al., 1993).In the United States, an annual average of only 68 deaths due <strong>to</strong> his<strong>to</strong>plasmosiswas recorded for 1952–1963, despite the high prevalence of the disease in endemicareas. This confirms that the disease is usually benign.In African his<strong>to</strong>plasmosis caused by var. duboisii, lesions occur most frequentlyon the skin, in subcutaneous tissue, <strong>and</strong> bones. Skin granulomas appear as nodulesor ulcerous or eczema<strong>to</strong>us lesions. Abscesses can be observed in subcutaneous tissue.Isolated or multiple lesions are found in osseous his<strong>to</strong>plasmosis, sometimesasymp<strong>to</strong>matically (Manson-Bahr <strong>and</strong> Apted, 1982). When the disease is progressive<strong>and</strong> severe, giant cell granulomas may form in many internal organs.Treatment of acute pulmonary his<strong>to</strong>plasmosis is justified only in severe <strong>and</strong> prolongedcases. Short-term treatment with intravenous amphotericin B for three orfour weeks is generally sufficient. Patients with disseminated his<strong>to</strong>plasmosis shouldusually be treated for a longer period with intravenous amphotericin B or oral ke<strong>to</strong>conazole(Loyd et al., 1990).


342 MYCOSESTwenty-seven AIDS patients with disseminated his<strong>to</strong>plasmosis were given oralitraconazole (200 mg per day <strong>to</strong> 24 patients <strong>and</strong> 400 mg per day <strong>to</strong> 3 patients) forsix months. Patients who were considered cured continued <strong>to</strong> take 100 mg per day.A <strong>to</strong>tal of 23 patients responded well <strong>to</strong> the treatment, three showed questionableresults, <strong>and</strong> one had a negative result (Negroni et al., 1992). Forty-two patients withAIDS <strong>and</strong> disseminated his<strong>to</strong>plasmosis who successfully completed treatment withamphotericin B for 4 <strong>to</strong> 12 weeks (15 mg/kg of body weight) were given itraconazole(200 mg twice a day) <strong>to</strong> prevent relapses, with satisfac<strong>to</strong>ry results (Wheatet al., 1993).The Disease in Animals: Dogs manifest clinical symp<strong>to</strong>ms most often but, as inman, most infections are asymp<strong>to</strong>matic. The primary respira<strong>to</strong>ry form of the diseasealmost always heals by encapsulation <strong>and</strong> calcification. In disseminated cases, thedogs lose weight <strong>and</strong> have persistent diarrhea, anorexia, <strong>and</strong> chronic cough;hepa<strong>to</strong>splenomegaly <strong>and</strong> lymphadenopathy may also be observed.Cats follow dogs in terms of frequency of clinical his<strong>to</strong>plasmosis. The symp<strong>to</strong>msof feline disseminated his<strong>to</strong>plasmosis are anemia, weight loss, lethargy, fever, <strong>and</strong>anorexia. In chest radiographs, the lungs of 7 of 12 cats indicated anomalies. Kittensone year of age or younger were most affected (Clinkenbeard et al., 1987).H. capsulatum has also been isolated from the intestinal contents <strong>and</strong> variousorgans of bats. High rates of reac<strong>to</strong>rs have been found in different domesticatedspecies (cattle, horses, sheep) in endemic areas <strong>and</strong> the agent has been isolated fromthe lymph nodes of dogs <strong>and</strong> cats as well as from a wild rodent (Proechimys guyanensis)<strong>and</strong> a sloth in Brazil. Birds are not susceptible <strong>to</strong> his<strong>to</strong>plasmosis, perhapsbecause their high body temperature does not allow the fungus <strong>to</strong> develop.Source of Infection <strong>and</strong> Mode of Transmission: The reservoir of the agent is thesoil, where it lives as a saprophyte. Its distribution in the soil is not uniform <strong>and</strong>depends on various fac<strong>to</strong>rs such as humidity, temperature, <strong>and</strong> others yet <strong>to</strong> be determined.Microfoci that have led <strong>to</strong> sporadic cases <strong>and</strong> epidemic outbreaks have usuallybeen associated with soils in which excreta from certain species of bird or batshave accumulated over some time. These excreta apparently allow the fungus <strong>to</strong>compete with other microorganisms in the soil, ensuring its survival. In contrast <strong>to</strong>birds, which are not infected by H. capsulatum <strong>and</strong> whose role in the epidemiologyis limited <strong>to</strong> the enabling function of their excreta, certain bat species, particularlyspecies that live in colonies, do become infected <strong>and</strong> eliminate the fungus in theirdroppings, thus contributing <strong>to</strong> its dissemination. Humans frequently becomeinfected when they visit caves, tunnels, <strong>and</strong> ab<strong>and</strong>oned mines <strong>and</strong> other placeswhere there are large populations of bats <strong>and</strong> much accumulated guano. Most infectionsin Mexico were due <strong>to</strong> exposure <strong>to</strong> bat droppings; cases occurred amongexplorers, <strong>to</strong>urists, spelunkers, geologists, biologists, <strong>and</strong> others entering suchplaces for work or study.Man <strong>and</strong> animals acquire the infection from the same source (the soil) throughinhalation. Microconidia of the fungus are the infecting element. The infection usuallystarts when natural foci are disturbed by activities that scatter the etiologic agentin the air, such as bulldozing, cleaning or demolishing rural structures (especiallyhenhouses), <strong>and</strong> visits <strong>to</strong> caves inhabited by bats.His<strong>to</strong>plasmosis occurs predominantly in rural areas, but outbreaks have alsooccurred among urban dwellers, particularly construction workers. This was the


HISTOPLASMOSIS 343case in outbreaks on the Indianapolis campus of Indiana University, where buildingdemolition <strong>and</strong> excavation led <strong>to</strong> many human cases (see the section on occurrencein man).In dogs, the disease appears more frequently in working <strong>and</strong> sporting breeds.Role of Animals in the Epidemiology of the Disease: Both man <strong>and</strong> animals areaccidental hosts of the etiologic agent <strong>and</strong> do not play a role in maintaining or transmittingthe infection. Only certain bat species are thought <strong>to</strong> play an active role indisseminating the infection, in addition <strong>to</strong> contributing <strong>to</strong> its development by meansof their droppings. However, further study is needed <strong>to</strong> assess the role of bats inspreading the agent from one roost <strong>to</strong> another, <strong>and</strong> <strong>to</strong> determine the susceptibility ofcertain species <strong>to</strong> his<strong>to</strong>plasmosis (Hoff <strong>and</strong> Bigler, 1981).Diagnosis: Labora<strong>to</strong>ry diagnosis can be performed through microscopic examinationof stained smears; immunofluorescence using clinical specimens such as sputum,ulcer exudate, <strong>and</strong> other materials; isolation in culture media; inoculation ofmice; <strong>and</strong> examination of his<strong>to</strong>pathologic sections (bone marrow, lung, liver, <strong>and</strong>spleen). In the acute pulmonary form, radiological findings of pulmonary infiltrates<strong>and</strong> hilar adenopathy, combined with data indicating that the patient comes from anendemic area <strong>and</strong> has symp<strong>to</strong>ms compatible with his<strong>to</strong>plasmosis, are enough <strong>to</strong>establish a presumptive diagnosis.Disseminated his<strong>to</strong>plasmosis is diagnosed by culturing the blood, bone marrow,urine, or other extrapulmonary tissues, or through biopsy <strong>and</strong> his<strong>to</strong>pathology. Severeacute, but not chronic, his<strong>to</strong>plasmosis can be diagnosed using a peripheral bloodsmear with Wright or Giemsa stain. Biopsy material from the liver or material fromoropharyngeal ulcers stained with silver methenamine yields good results (Loyd etal., 1990).The his<strong>to</strong>plasmin test is administered like the tuberculin test <strong>and</strong> read at 24 <strong>and</strong>48 hours. Sensitivity is established one <strong>to</strong> two months after infection <strong>and</strong> lasts formany years. Although this test is extremely value for epidemiological research, itsusefulness is limited in clinical diagnosis. It is advisable <strong>to</strong> administer the test<strong>to</strong>gether with the coccidioidin <strong>and</strong> blas<strong>to</strong>mycin tests because of cross-reactions. Anegative test in a patient can indicate that the infection is recent or that the diseasehas a different etiology.Serological tests (complement fixation, immunodiffusion, radioimmunoassay,enzyme immunoassay, precipitation, latex agglutination) are useful for diagnosis butnot very sensitive or specific. Tests for blas<strong>to</strong>mycosis <strong>and</strong> coccidioidomycosisshould be performed at the same time. It should be kept in mind that a his<strong>to</strong>plasmintest can produce antibodies; thus, it is recommended that the blood sample be takenwhen the allergy test is conducted. It is expected that a test that detects H. capsulatumantigen in serum <strong>and</strong> urine will give more specific results (Wheat et al., 1986).Control: The principal protection measure consists of reducing people’s exposure<strong>to</strong> dust by spraying with a 3%–5% formalin solution on the ground when cleaninghenhouses or other potentially contaminated sites. The use of protective masks hasbeen recommended. Control of natural foci is difficult. During one outbreak, it waspossible <strong>to</strong> eradicate the fungus from its natural foci by spraying the soil with formol.


344 MYCOSESBibliographyAjello, L. Comparative ecology of respira<strong>to</strong>ry mycotic disease agents. Bact Rev 31:6–24, 1967.Ajello, L., W. Kaplan. Systemic mycoses. In: S<strong>to</strong>enner, H., W. Kaplan, M. Torten, eds.Section A, Vol 2: CRC H<strong>and</strong>book Series in Zoonoses. Boca Ra<strong>to</strong>n: CRC Press; 1980.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Borelli, D. Prevalence of systemic mycoses in Latin America. In: Pan American HealthOrganization. Proceedings: International Symposium on Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>;1970. (Scientific Publication 205).Clinkenbeard, K.D., R.L. Cowell, R.D. Tyler. Disseminated his<strong>to</strong>plasmosis in cats: 12 cases(1981–1986). J Am Vet Med Assoc 190:1445–1448, 1987.Cole, C.R., R.L. Farrell, D.M. Chamberlain, et al. His<strong>to</strong>plasmosis in animals. J Am Vet MedAssoc 122:471–473, 1953.Conces, D.J., S.M. S<strong>to</strong>ckberger, R.D. Tarver, I.J. Wheat. Disseminated his<strong>to</strong>plasmosis inAIDS: Findings on chest radiographs. Am J Roentgenol 160:15–19, 1993.Coulanges, P. L’his<strong>to</strong>plasmose a gr<strong>and</strong>es formes (H. duboisii) a Madagascar (A propos de 3cas). Arch Inst Pasteur Madagascar 56:169–174, 1989.González Menocal, I., M. Suárez Menéndez, L. Pérez González, J. Díaz Rodríguez. Estudioclínico-epidemiológico de un brote de his<strong>to</strong>plasmosis pulmonar en el Municipio de Morón.Rev Cubana Hig Epidemiol 28:179–183, 1990.Hamil<strong>to</strong>n, A.J., M.A. Bartholomew, L. Fenelon, et al. Preparation of monoclonal antibodiesthat differentiate between His<strong>to</strong>plasma capsulatum variant capsulatum <strong>and</strong> H. capsulatumvariant duboisii. Trans Roy Soc Trop Med Hyg 84:425–428, 1990.Hoff, G.L., W.J. Bigler. The role of bats in the propagation <strong>and</strong> spread of his<strong>to</strong>plasmosis: Areview. J Wild Dis 17:191–196, 1981.Johnson, P.C., N. Khardori, A.F. Naijar, et al. Progressive disseminated his<strong>to</strong>plasmosis inpatients with acquired immunodeficiency syndrome. Am J Med 85:152–158, 1988.Kaplan, W. Epidemiology of the principal systemic mycoses of man <strong>and</strong> lower animals <strong>and</strong>the ecology of their etiologic agents. J Am Vet Med Assoc 163:1043–1047, 1973.Loyd, J.E., R.M. Des Prez, R.A. Goodwin, Jr. His<strong>to</strong>plasma capsulatum. In M<strong>and</strong>ell, G.L.,R.G. Douglas, Jr., J.E. Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed.New York: Churchill Livings<strong>to</strong>ne Inc.; 1990.Manson-Bahr, P.E.C., F.I.C. Apted. Manson’s Tropical Diseases. 18th ed. London: Baillière-Tindall; 1982.Menges, R.W., R.T. Habermann, L.A. Selby, H.R. Ellis, R.F. Behlow, C.D. Smith. A review<strong>and</strong> recent findings of his<strong>to</strong>plasmosis in animals. Vet Med 58:334–338, 1963.Mir<strong>and</strong>a Novales, M.G., F. Solórzano San<strong>to</strong>s, H. Díaz Ponce, et al. His<strong>to</strong>plasmosis diseminadaen pediatría. Bol Med Hosp Infant Mex 50:272–275, 1993.Negroni, P. His<strong>to</strong>plasmosis: Diagnosis <strong>and</strong> Treatment. Springfield: Thomas; 1965.Negroni, P. Micosis cutáneas y viscerales. 5.ª ed. Buenos Aires: López; 1972.Negroni, R., A. Taborda, A.M. Robles, A. Archevala. Itraconazole in the treatment of his<strong>to</strong>plasmosisassociated with AIDS. Mycoses 35:281–287, 1992.Organización Panamericana de la Salud (OPS). His<strong>to</strong>plasmosis en México, 1979–1980. BolEpidemiol 2:12–13, 1981.Sanger, V.L. His<strong>to</strong>plasmosis. In: Davis, J.W., L.H. Karstad, D.O. Trainer, eds. InfectiousDiseases of Wild Mammals. Ames: Iowa State University Press; 1970.Schlech, W.F., L.J. Wheat, J.L. Ho, M.L.V. French, R.J. Weeks, R.B. Kohler, et al. Recurrenturban his<strong>to</strong>plasmosis, Indianapolis, Indiana. 1980–1981. Am J Epidemiol 118:301–302, 1983.Selby, L.A. His<strong>to</strong>plasmosis. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger, eds.Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.


MYCETOMA 345Sweany, H.C., ed. His<strong>to</strong>plasmosis. Springfield, Illinois: Thomas; 1960.Waldman, R.J., A.C. Engl<strong>and</strong>, R. Tauxe, T. Kline, R.J. Weeks, L.A. Ajello, et al. A winteroutbreak of acute his<strong>to</strong>plasmosis in northern Michigan. Am J Epidemiol 117:68–75, 1983.Wheat, J., R. Hafner, M. Wulfsohn, et al. Prevention of relapse of his<strong>to</strong>plasmosis with itraconazolein patients with acquired inmunodeficiency syndrome. The National Institute ofAllergy <strong>and</strong> Infectious Diseases. Clinical Trials <strong>and</strong> Mycoses Study Group Collabora<strong>to</strong>rs. AnnIntern Med 118:610–616, 1993.Wheat, L.J., R.B. Kohler, R.P. Tewari. Diagnosis of disseminated his<strong>to</strong>plasmosis by detectionof His<strong>to</strong>plasma capsulatum antigen in serum <strong>and</strong> urine specimens. N Engl J Med314:83–88, 1986.MYCETOMAICD-10 B47.0 eumyce<strong>to</strong>ma; B47.1 actinomyce<strong>to</strong>maSynonyms: Maduromycosis, Madura foot, maduromycotic myce<strong>to</strong>ma, eumycoticmyce<strong>to</strong>ma, actinomyce<strong>to</strong>ma.Etiology: Myce<strong>to</strong>mas may be caused by many species of fungi (eumyce<strong>to</strong>ma) orby bacterial agents (actinomyce<strong>to</strong>ma). The principal agents of eumyce<strong>to</strong>ma areMadurella myce<strong>to</strong>matis, M. grisea, Lep<strong>to</strong>sphaeria senegalensis (all of which produceblack granules), Pseudallescheria (Petriellidium, Allescheria) boydii,various speciesof Acremonium (white or yellow granules), Exophiala jeanselmei, <strong>and</strong> other speciesof fungi. Actinomyce<strong>to</strong>mas are caused by Nocardia brasiliensis, N. asteroides, <strong>and</strong>N. otitidiscaviarum, Strep<strong>to</strong>myces somaliensis, Actinomadura madurae, <strong>and</strong> A. pelletieri.The principal agents of animal myce<strong>to</strong>mas are P. boydii, Curvularia geniculata,Cochliolobus spicifer, Acremonium spp., <strong>and</strong> Madurella grisea.Both the fungi <strong>and</strong> the actinomycetes are soil saprophytes that accidentally enterthe host’s tissues, where they form granules (colonies). Eumyce<strong>to</strong>ma granules containthick hyphae whereas actinomyce<strong>to</strong>ma granules contain fine filaments.Geographic Distribution: The agents of maduromycosis are distributed worldwidebut occur primarily in the tropics. In tropical areas of Africa <strong>and</strong> India, infectionis most frequently caused by Madurella myce<strong>to</strong>matis <strong>and</strong> Strep<strong>to</strong>myces somaliensis.In Mexico, Central America, <strong>and</strong> South America, myce<strong>to</strong>mas are causedprimarily by Nocardia brasiliensis <strong>and</strong> Actinomadura madurae; in Canada <strong>and</strong> theUnited States, they are caused primarily by Pseudallescheria boydii; <strong>and</strong> in Japanthey are due <strong>to</strong> Nocardia asteroides (Mahgoub, 1990).Occurrence in Man: Infrequent. It is more <strong>common</strong> in tropical <strong>and</strong> subtropicalzones, particularly where people go barefoot.Most cases occur in Africa. In Sudan, 1,231 patients required hospitalization in atwo-<strong>and</strong>-a-half-year period. In many African countries, such as Cameroon, Chad,Kenya, Mauritania, Niger, Senegal, Somalia, <strong>and</strong> Sudan, myce<strong>to</strong>ma is considered


346 MYCOSESthe most frequent deep mycosis (Develoux et al., 1988). The responsible agents inAfrica vary by geographic area. In India, myce<strong>to</strong>ma is endemic in many areas. In theAmericas, it occurs most frequently in Mexico <strong>and</strong> Central America (due primarily<strong>to</strong> Nocardia brasiliensis) (Manson-Bahr <strong>and</strong> Apted, 1982). In São Paulo (Brazil)there were 154 cases between 1944 <strong>and</strong> 1978; 73.4% of these were actinomyce<strong>to</strong>mas<strong>and</strong> 26.6% were eumyce<strong>to</strong>mas. In Niger, men are infected more frequentlythan women (4:1). The disease occurs in rural areas.Occurrence in Animals: Rare.The Disease in Man: Myce<strong>to</strong>ma is a slow-developing, chronic infection that usuallylocalizes on the foot, the lower leg, sometimes the h<strong>and</strong>, <strong>and</strong> rarely on someother part of the body. The incubation period is several months from the time ofinoculation. The lesion may begin as a papule, nodule, or abscess. The myce<strong>to</strong>maspreads <strong>to</strong> deep tissue <strong>and</strong> the foot (or h<strong>and</strong>) swells <strong>to</strong> two or three times its normalsize. Numerous small abscesses form, as well as fistulous tracks in the subcutaneoustissue that branch out <strong>to</strong> the tendons <strong>and</strong> may reach the bones. Pus discharged <strong>to</strong> thesurface contains characteristic granules (microcolonies) that may be white oranother color depending on the causal agent. The skin does not lose sensitivity nordoes the patient generally feel any pain. Actinomyce<strong>to</strong>mas almost always respond <strong>to</strong>treatment with antibacterial antibiotics (strep<strong>to</strong>mycin, co-trimoxazole), but eumyce<strong>to</strong>masare quite resistant (ke<strong>to</strong>conazole, myconazole) <strong>and</strong> often lead <strong>to</strong> amputation.Oral dapsone is preferred for cases of Actinomadura madurae. The same treatmentis recommended for patients affected by Strep<strong>to</strong>myces somaliensis but should bechanged <strong>to</strong> trimethoprim/sulfamethoxazole tablets if no improvement is seen afterone month. The latter treatment is also used for infections caused by Nocardia spp.(Mahgoub, 1990).The Disease in Animals: Almost all confirmed cases have occurred in theUnited States. In animals (dogs, cats, horses), eumyce<strong>to</strong>mas are localized in thefeet, lymph nodes, abdominal cavity, <strong>and</strong> other areas of the body. The most <strong>common</strong>agents of eumyce<strong>to</strong>ma in animals are Curvularia geniculata <strong>and</strong>Pseudallescheria boydii. Myce<strong>to</strong>mas are frequently preceded by traumas. Intraabdominalinfections have been described in dogs in association with ovariohysterec<strong>to</strong>miesor a surgical incision that had opened up, with surgery occurring twoyears prior <strong>to</strong> the appearance of clinical symp<strong>to</strong>ms. Lesions seen in animals aresimilar <strong>to</strong> those in humans. They generally start with a small subcutaneous nodulethat grows gradually for months or years. They may become deeper <strong>and</strong> destroyunderlying tissues (McEntee, 1987).Cases of kera<strong>to</strong>mycosis <strong>and</strong> other ophthalmic conditions due <strong>to</strong> Pseudallescheriaboydii have been described in humans as well as horses <strong>and</strong> dogs (Friedmanet al., 1989).Source of Infection <strong>and</strong> Mode of Transmission: The etiologic agents of this diseaseare saprophytes in soil <strong>and</strong> vegetation. The fungus is introduced in<strong>to</strong> subcutaneoustissue of humans <strong>and</strong> animals through wounds. Contaminated thorns or splintersmay be the immediate source of the infection. In animals, there are cases ofpost-operative wounds being infected by P. boydii.Role of Animals in the Epidemiology of the Disease: None.


MYCETOMA 347Diagnosis: Microscopic examination of pus or material from curettage or biopsycan distinguish eumyce<strong>to</strong>ma granules from actinomyce<strong>to</strong>ma (nocardiosis) granules.The agent is identified through isolation in culture media such as Lowenstein-Jensenmedium for actinomyce<strong>to</strong>ma granules <strong>and</strong> blood agar for eumyce<strong>to</strong>ma granules.Sabouraud’s agar is used for subculturing with antimicrobial antibiotics. It is recommendedthat biopsy material, rather than material from the fistulae, be used <strong>to</strong>obtain the granules aseptically (Mahgoub, 1990). It is advisable <strong>to</strong> determine theagent’s sensitivity <strong>to</strong> different medications in order <strong>to</strong> ensure correct treatment.In a study conducted in Sudan, specific diagnosis was achieved for 78% of thespecimens by using his<strong>to</strong>logic methods, <strong>and</strong> for 82% of the cases by immunodiffusion(Mahgoub, 1975). The choice of strains for serological testing is very important.Control: Humans can avoid becoming infected by wearing shoes.BibliographyBenenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Brodey, R.S., H.F. Schryver, M.J. Deubler, W. Kaplan, L. Ajello. Myce<strong>to</strong>ma in a dog. J AmVet Med Assoc 151:442–451, 1967.Conant, N.F. Medical mycology. In: Dubos, R.J., J.G. Hirsch, eds. Bacterial <strong>and</strong> ClinicalMycology. 2nd ed. Philadelphia: Saunders; 1963.Conant, N.F. Medical mycology. In: Dubos, R.J., J.G. Hirsch, eds. Bacterial <strong>and</strong> MycoticInfections of Man. 4th ed. Philadelphia: Lippincott; 1965.Develoux, M., J. Audoin, J. Tregner, et al. Myce<strong>to</strong>ma in the Republic of Niger: Clinical features<strong>and</strong> epidemiology. Am J Med Trop Hyg 38:386–390, 1988.Friedman, D., J.V. Schoster, J.P. Pickett, et al. Pseudallescheria boydii kera<strong>to</strong>mycosis in ahorse. J Am Vet Med Assoc 195:616–618, 1989.Jang, S.S., J.A. Popp. Eumycotic myce<strong>to</strong>ma in a dog caused by Allescheria boydii. J AmVet Med Assoc 157:1071–1076, 1970.Mahgoub, E.S. Serologic diagnosis of myce<strong>to</strong>ma. In: Pan American Health Organization.Proceedings of the Third International Conference on the Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>;1975. (Scientific Publication 304).Mahgoub, E.S. Agents of myce<strong>to</strong>ma. In: M<strong>and</strong>ell, G.L., R.G. Douglas, Jr., J.E. Bennett,eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York: Churchill Livings<strong>to</strong>neInc.; 1990.Manson-Bahr, P.E.C., F.I.C. Apted. Manson’s Tropical Diseases. 18th ed. London:Baillière-Tindall; 1982.McEntee, M. Eumycotic myce<strong>to</strong>ma: Review <strong>and</strong> report of a cutaneous lesion caused byPseudallescheria boydii in a horse. J Am Vet Med Assoc 191:1459–1461, 1987.Segretain, G., F. Mariar. Myce<strong>to</strong>ma. In:Warren, K.S., A.A.F. Mahmoud, eds. Tropical <strong>and</strong>Geographical Medicine. New York: McGraw-Hill; 1984.


348 MYCOSESPROTOTHECOSISSynonyms: Algal infections.Etiology: In recent years, mycologists have called attention <strong>to</strong> infections inhumans <strong>and</strong> animals caused by microorganisms of the genus Pro<strong>to</strong>theca, the nature<strong>and</strong> taxonomy of which have not yet been clearly defined. Most authors believe theyare unicellular algae, but others describe them as algae-like fungi.The cells of Pro<strong>to</strong>theca spp. are round or oval <strong>and</strong> measure between 2 <strong>and</strong> 16microns in diameter. The species of interest are P. wickerhamii <strong>and</strong> P. zopfii. Thesemicroorganisms reproduce asexually. Hyaline cells, called sporangia when mature,produce from 2 <strong>to</strong> 20 endospores in their interior that increase in volume <strong>and</strong> repeatthe reproductive cycle when they reach maturity.Geographic Distribution: The agents are distributed worldwide.Occurrence in Man: Slightly more than 30 cases of pro<strong>to</strong>thecosis have beendescribed, 60% of them in men. With the exception of one case due <strong>to</strong> P. zopfii, thecausal agent in all other cases in which the species was identified was P. wickerhamii.Recently, an infection caused by green algae was described (Jones, 1983).Occurrence in Animals: Pro<strong>to</strong>thecosis occurs in many animal species, but aboveall in cattle <strong>and</strong> dogs. Numerous isolations have been recorded (McDonald et al.,1984). Most infections are due <strong>to</strong> P. zopfii. Occurrence is sporadic. Nevertheless, 23infected animals were found in one dairy herd of 90 cows.Mastitis caused by P. zopfii in cattle is more frequent than was formerly thought.In the United States, there were 400 reported cases in 1982 in New York State alone(Mayberry, 1984, cited in Pore et al., 1987). In Australia, mastitis due <strong>to</strong> P. zopfiiwas diagnosed in 17 of the 120 cows in a herd (Hodges et al., 1985). There were 10cases in a herd of 192 cows in Denmark <strong>and</strong> 5 cases in a herd of 130 cows in GreatBritain (Pore et al., 1987).The Disease in Man: The incubation period is unknown. Pro<strong>to</strong>thecosis manifestsitself in two principal clinical forms (Kaplan, 1978). One is progressive ulcerativeor verrucous lesions of the cutaneous <strong>and</strong> subcutaneous tissue on exposed skin. Theother is chronic olecranon bursitis, with pain <strong>and</strong> swelling. In one case of dissemination,intraperi<strong>to</strong>neal <strong>and</strong> facial nodules were observed.Treatment consists of surgical excision of the lesion. Antibacterial medications areineffective. Of the antimycotics, amphotericin B has produced satisfac<strong>to</strong>ry results.The Disease in Animals: The predominant form of pro<strong>to</strong>thecosis in cattle is mastitis,which at times may affect all four quarters of the udder. Temperature <strong>and</strong>appetite may remain normal. Inflammation of the udder is mild in comparison withbacterial mastitis, but it is invasive <strong>and</strong> chronic. The etiological agent causes pyogranulomasin the mammary gl<strong>and</strong> <strong>and</strong> the regional lymph nodes (Pore et al., 1987).Milk production in the affected quarter diminishes, <strong>and</strong> small clots may be found inthe milk. The disease was reproduced experimentally using a small number of P.zopfii (McDonald et al., 1984).Pro<strong>to</strong>thecosis in dogs is usually a systemic disease, with dissemination of theinfection <strong>to</strong> many internal organs. The severity of the disease varies according <strong>to</strong> the


PROTOTHECOSIS 349organs affected. Weakness <strong>and</strong> weight loss were observed in all cases of dissemination(Kaplan, 1978).Approximately one-half of the cases in dogs are caused by P. wickerhamii <strong>and</strong> theother half by P. zopfii (Dillberger et al., 1988). Other animal species in which pro<strong>to</strong>thecosishas been diagnosed are Atlantic salmon <strong>and</strong> cats. In salmon, P. salmoniscauses a disseminated <strong>and</strong> fatal disease (Gentles <strong>and</strong> Bond, 1977). The clinical manifestationof pro<strong>to</strong>thecosis in cats more closely resembles the cutaneous disease inhumans <strong>and</strong> does not tend <strong>to</strong> disseminate as it does in dogs. The infection in cats iscaused by P. wickerhamii (Dillberger et al., 1988).Source of Infection <strong>and</strong> Mode of Transmission: Pro<strong>to</strong>theca spp. <strong>and</strong> greenalgae are saprophytes found in nature, primarily in stagnant or slow-moving waters.Humans acquire the infection, possibly through skin lesions, when they come in<strong>to</strong>contact with contaminated water or other habitats of these agents. The profusion ofthese agents in the environment, as well as the few cases described in humans, indicatethat they are not very virulent <strong>and</strong> that lowered host resistance is required forthem <strong>to</strong> act as pathogens. In fact, five of nine patients with cutaneous or subcutaneouspro<strong>to</strong>thecosis had a preexisting or intercurrent disease. Similarly, seven ofeight patients with the olecranon bursitis form had previously sustained a trauma <strong>to</strong>the elbow (Kaplan, 1978). Cattle contract mastitis caused by P. zopfii in the environmentitself; the portal is probably the teat. P. zopfii is abundant in dairies, in cowfeces as well as in drinking troughs, feed, <strong>and</strong> mud. A study conducted on variousdairy cows, some with mastitis caused by Pro<strong>to</strong>theca <strong>and</strong> others without any his<strong>to</strong>ryof the disease, isolated the agent (94% P. zopfii <strong>and</strong> 6% P. wickerhamii) in 48(25.3%) of 190 samples (Anderson <strong>and</strong> Walker, 1988). Little is known of the predisposingconditions in dogs, which almost always manifest systemic pro<strong>to</strong>thecosis.In cattle, the retropharyngeal <strong>and</strong> m<strong>and</strong>ibular lymph nodes affected by green algaeindicate that the infection is possibly contracted by ingestion of contaminated water.The few cases described in cattle <strong>and</strong> sheep suggest that these species are not verysusceptible <strong>to</strong> green algal infection.Diagnosis: Special stains such as Gomori, Gridley, <strong>and</strong> PAS (periodic acid-Schiff) applied <strong>to</strong> his<strong>to</strong>logical sections from affected tissues permit detection ofPro<strong>to</strong>theca in all developmental stages. To determine the species, cultures or theimmunofluorescence test with species-specific reagents must be used. The immunofluorescencetechnique can be used for cultures as well as for his<strong>to</strong>logical sectionsstained with hema<strong>to</strong>xylin-eosin, but not for those stained with the methods mentionedabove.Control: Treatment of underlying conditions or <strong>diseases</strong> in humans.BibliographyAnderson, K.L., R.L. Walker. Sources of Pro<strong>to</strong>theca spp. in a dairy herd environment. J AmVet Med Assoc 193:553–556, 1988.Dillberger, J.E., B. Homer, D. Daubert, N.H. Altman. Pro<strong>to</strong>thecosis in two cats. J Am VetMed Assoc 192:1557–1559, 1988.Gentles, J.C., P.M. Bond. Pro<strong>to</strong>thecosis of Atlantic salmon. Sabouraudia 15:133–139, 1977.


350 MYCOSESHodges, R.T., J.T.S. Holl<strong>and</strong>, F.J.A. Neilson, N.M. Wallace. Pro<strong>to</strong>theca zopfii mastitis in aherd of dairy cows. N Z Vet J 33:108–111, 1985.Jones, J.W., H.W. McFadden, F.W. Ch<strong>and</strong>ler, W. Kaplan, D.H. Conner. Green algal infectionin a human. Am J Clin Pathol 80:102–107, 1983.Kaplan, W. Pro<strong>to</strong>thecosis <strong>and</strong> infections caused by morphologically similar green algae. In:Pan American Health Organization. Proceedings of the Fourth International Conferenceon Mycoses: The Black <strong>and</strong> White Yeasts. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1978. (ScientificPublication 356).Kaplan, W., F.W. Ch<strong>and</strong>ler, C. Choudary, P.K. Ramach<strong>and</strong>ran. Disseminated unicellulargreen algal infection in two sheep in India. Am J Trop Med Hyg 32:405–411, 1983.Mayberry, D. Colorless alga can pollute water, cause mastitis. Agri Res March: 4–5, 1984.Cited in: Pore, R.S., et al. Occurrence of Pro<strong>to</strong>theca zopfii, a mastitis pathogen, in milk. VetMicrobiol 15:315–323, 1987.McDonald, J.S., J.L. Richard, N.F. Cheville. Natural <strong>and</strong> experimental bovine intramammaryinfection with Pro<strong>to</strong>theca zopfii. Am J Vet Res 45:592–595, 1984.Pore, R.S., T.A. Shahan, M.D. Pore, R. Blauwiekel. Occurrence of Pro<strong>to</strong>theca zopfii,a mastitispathogen, in milk. Vet Microbiol 15:315–323, 1987.Rogers, R.J., M.D. Connole, J. Nor<strong>to</strong>n, A. Thomas, P.W. Ladds, J. Dickson. Lymphadenitisof cattle due <strong>to</strong> infection with green algae. J Comp Pathol 90:1–9, 1980.RHINOSPORIDIOSISICD-10 B48.1Etiology: Rhinosporidium seeberi, a fungus that in tissue forms sporangia containinga large number of sporangiospores. Its environmental habitat is unknown <strong>and</strong>its taxonomy uncertain.Geographic Distribution: The disease has been confirmed in the Americas, Asia(endemic zones in India <strong>and</strong> Sri Lanka), Africa, Europe, Australia, <strong>and</strong> NewZeal<strong>and</strong>.Occurrence in Man <strong>and</strong> Animals: The disease is rare throughout the world. Up<strong>to</strong> 1970, data from Latin America show 108 cases in humans. Most occurred inParaguay (56), Brazil (13), <strong>and</strong> Venezuela (13) (Mayorga, 1970). According <strong>to</strong> morerecent data, more than 50 cases have occurred in Venezuela, mainly in the states ofBarinas <strong>and</strong> Portuguesa. In addition <strong>to</strong> the Latin American countries already cited,the disease has been confirmed in Argentina <strong>and</strong> Cuba. In the United States, some30 cases have been recorded, primarily in the south. Five cases have been reportedin Trinidad (Raju <strong>and</strong> Jamalabadi, 1983), four of them affecting the conjunctiva.Most of the cases in Africa were recorded in Ug<strong>and</strong>a. A retrospective study(1948–1986) of 91,000 biopsies was conducted at the Central Hospital of Mapu<strong>to</strong>,Mozambique; rhinosporidiosis was diagnosed in 33 (0.036%) (Moreira Díaz et al.,1989). Some 1,000 cases have occurred in India <strong>and</strong> Sri Lanka, <strong>and</strong> 72 occurred inIran over a 30-year period.


RHINOSPORIDIOSIS 351The disease is seen mostly in children <strong>and</strong> young people, predominantly in males(Mahapatra, 1984).Rhinosporidiosis in animals occurs in cattle, horses, dogs, cats, <strong>and</strong> geese. Morethan 90% of the cases occur in males (Carter <strong>and</strong> Chengappa, 1991). The diseaseoccurs sporadically, as it does in humans. An unusual case occurred in a province innorthern Argentina where an outbreak was described in a herd of cattle that was keptin a flooded field for two years. Twenty-four percent of the animals examined hadpolyps (Luciani <strong>and</strong> Toledo, 1989).The Disease in Man <strong>and</strong> Animals: Rhinosporidiosis is characterized by pedunculatedor sessile polyps on the mucous membranes, particularly of the nose <strong>and</strong>eyes. The polyps are soft, lobular, <strong>and</strong> reddish with small white spots (the sporangia).These excrescences are not painful but they do bleed easily. In humans, thesegranuloma<strong>to</strong>us formations can also be found in the pharynx, larynx, ear, vagina,penis, rectum, <strong>and</strong> on the skin. Cases of dissemination <strong>to</strong> internal organs are rare.The clinical picture in animals consists of a chronic polypoid inflammation thatmay cause respira<strong>to</strong>ry difficulty <strong>and</strong> sneezing if the disease lodges in the nasalmucosa <strong>and</strong> if the lesion is sufficiently large. Another <strong>common</strong> symp<strong>to</strong>m is epistaxis.Treatment for humans <strong>and</strong> animals consists of surgical excision of the polyp.Recurrence is rare. Successful treatment with dapsone has been described in threepatients (Job et al., 1993).Source of Infection <strong>and</strong> Mode of Transmission: The natural habitat of the agentis unknown. It is suspected that the infection enters the body with soil particlesthrough lesions of the mucous membranes. Those affected almost always live inrural areas, thus the assumption that the agent lives in the soil. In India <strong>and</strong> SriLanka, where most cases have been recorded, the source of infection has been associatedwith stagnant waters, but it has not yet been possible <strong>to</strong> demonstrate the presenceof the fungus in such waters or in aquatic animals. The route of infection <strong>and</strong>the mode of transmission are also unknown.Role of Animals in the Epidemiology of the Disease: Rhinosporidiosis is a disease<strong>common</strong> <strong>to</strong> humans <strong>and</strong> animals, contracted from an as yet unknown environmentalsource. It is not transmitted from one individual <strong>to</strong> another.Diagnosis: Since the fungus cannot be cultured, diagnosis depends on the clinicalappearance of the lesions <strong>and</strong> demonstration of the agent’s presence in tissues.Best results are obtained by using stained his<strong>to</strong>logical preparations.Control: No practical control measures are available.BibliographyAjello, L., L.K. Georg, W. Kaplan, L. Kaufman. Labora<strong>to</strong>ry Manual for MedicalMycology. Washing<strong>to</strong>n, D.C.: U.S. Government Printing Office; 1963. (Public Health ServicePublication 994).Carter, G.R., M.M. Chengappa. Essentials of Veterinary Bacteriology <strong>and</strong> Mycology. 4thed. Philadelphia: Lea & Febiger; 1991.Job, A., S. VanKateswaran, M. Mathan, et al. Medical therapy of rhinosporidiosis with dapsone.J Laryngol O<strong>to</strong>l 107:809–812, 1993.


352 MYCOSESLuciani, C.A. H.O. Toledo. Rhinosporidium seeberi en bovinos criollos (Bos taurus). VetArg 6(57):451–455, 1989.Mahapatra, L.N. Rhinosporidiosis. In: Warren, K.S., A.A.F. Mahmoud, eds. Tropical <strong>and</strong>Geographical Medicine. New York: McGraw-Hill; 1984.Mayorga, R. Prevalence of subcutaneous mycoses in Latin America. In: Pan AmericanHealth Organization. Proceedings: International Symposium on Mycoses. Washing<strong>to</strong>n, D.C.:<strong>PAHO</strong>; 1970. (Scientific Publication 205).Moreira Díaz, E.E., B. Milán Batista, C.E. Mayor González, H. Yokoyama.Rinosporidiosis: estudio de 33 casos diagnosticados por biopsias en el Hospital Central deMapu<strong>to</strong>, desde 1944 hasta 1986. Rev Cubana Med Trop 41:461–472, 1989.Negroni, P. Micosis cutáneas y viscerales. 5.ª ed. Buenos Aires: López; 1972.Raju, G.C., M.H. Jamalabadi. Rhinosporidiosis in Trinidad. Trop Geogr Med 35:257–258, 1983.Sauerteig, E.M. Rhinosporidiosis in Barinas, Venezuela. In: Pan American HealthOrganization. Proceedings of the Fifth International Conference on the Mycoses: Superficial,Cutaneous, <strong>and</strong> Subcutaneous Infections. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1980. (ScientificPublication 396).Utz, J.P. The mycoses. In: Beeson, P.B., W. McDermott, J.B. Wyngaarden, eds. CecilTextbook of Medicine. 15th ed. Philadelphia: Saunders; 1979.SPOROTRICHOSISICD-10 B42.0 pulmonary sporotrichosis; B42.1 lymphocutaneous sporotrichosis;B42.7 disseminated sporotrichosis; B42.8 other forms of sporotrichosisEtiology: Sporothrix schenckii (Sporotrichum schenckii, Sporotrichum beurmanni),a saprophytic fungus that lives in soil, plants, wood, <strong>and</strong> decaying vegetation.S. schenckii is a dimorphic fungus that occurs in a mycelial form in nature <strong>and</strong> ayeast form in infected animal tissues or on enriched culture media (such as bloodagar) at 37°C. The latter form generally produces multiple buds <strong>and</strong> occasionally asingle bud.Geographic Distribution: Worldwide; more <strong>common</strong> in tropical regions.Occurrence in Man: Sporadic; its frequency varies from region <strong>to</strong> region. Thedisease has been confirmed in all Latin American countries except Bolivia, Chile,<strong>and</strong> Nicaragua. It is more frequent in Asia, Brazil, the Central American countries,Mexico, South Africa, <strong>and</strong> Zimbabwe than in other countries. Although it is a relativelyrare disease, an epidemic affecting 3,000 workers was recorded in SouthAfrican gold mines. One group of cases also occurred in the United States amongforestry workers who contracted the disease while planting pine trees, <strong>and</strong> anothergroup of cases occurred among students who came in contact with contaminatedbricks (Mitchell, 1983). The largest outbreak in the United States, encompassing 15states, occurred in the spring of 1988 <strong>and</strong> affected 84 people. The outbreak was due<strong>to</strong> S. schenckii in sphagnum moss that was used <strong>to</strong> pack young plants for shipment


SPOROTRICHOSIS 353(Coles et al., 1992). In the area around Ayerza Lagoon in Guatemala, 53 cases wereseen between 1971 <strong>and</strong> 1975 (Mayorga et al., 1979). Results of skin hypersensitivitytests using S. schenckii <strong>and</strong> Cera<strong>to</strong>cystis stenoceras (a closely related species)antigens indicated that asymp<strong>to</strong>matic infection is probably frequent among peoplewho work with plants. The study done in the Ayerza Lagoon region (Mayorga et al.,1979) found that cutaneous hypersensitivity was at least 10 times higher amonglocal inhabitants than among residents of Guatemala City.The disease is much more frequent in males than in females.Occurrence in Animals: Occasional. Horses are the most frequently affected.Cases have been recorded in dogs, cats, rodents, cattle, swine, camels, birds, <strong>and</strong>wild animals.The Disease in Man: The incubation period can range from three weeks <strong>to</strong> threemonths. The most <strong>common</strong> clinical form is the cutaneous form; it begins with a noduleor pustule at the point where broken skin allowed inoculation. The primarylesion is usually located on exposed extremities. The infection may remain confined<strong>to</strong> the entry site or may eventually spread <strong>and</strong> produce subcutaneous nodules alongthe enlarged lymph nodes. The nodules may ulcerate, <strong>and</strong> a gray or yellowish pusappears. The patient’s general state of health is usually not affected. There are alsovegetative <strong>and</strong> verrucous dermal <strong>and</strong> epidermal forms.Disseminated forms, which are rare, may give rise <strong>to</strong> localizations in differen<strong>to</strong>rgans, especially the bones <strong>and</strong> joints (80% of extracutaneous forms) as well as themouth, nose, kidneys, <strong>and</strong> subcutaneous tissue over large areas of the body. Of morethan 3,000 miners who contracted cutaneous sporotrichosis, only five developedsystemic infections <strong>and</strong> none developed the pulmonary form (Lurie, 1962). Someresearchers have concluded that dissemination occurs via the bloodstream or thelymphatic system from the inoculation site on the skin, while others believe that aprimary focus in the lungs is involved.Pulmonary sporotrichosis, a rare form of the disease, results from inhalation ofthe fungus. Its course may be acute, but in general it is chronic <strong>and</strong> can be confusedwith tuberculosis. The number of cases described is probably less than 90, <strong>and</strong> mostpatients lived in states bordering the Mississippi <strong>and</strong> Missouri rivers in the UnitedStates. Many of them had underlying <strong>diseases</strong>, such as alcoholism <strong>and</strong> tuberculosis.The most <strong>common</strong> symp<strong>to</strong>ms are cough (69%), expec<strong>to</strong>ration (59%), dyspnea, pleuriticpain, <strong>and</strong> hemoptysis. Patients frequently complain of weight loss, fatigue, <strong>and</strong>a slight rise in body temperature. The most frequent lesion in the lungs occurs in theupper lobe, <strong>and</strong> radiography shows cavitation, surrounded by parenchyma<strong>to</strong>us densities(Pluss <strong>and</strong> Opal, 1986).Oral potassium iodide may be used <strong>to</strong> treat the cutaneous form. Extracutaneouscases have been treated successfully with ke<strong>to</strong>conazole <strong>and</strong> itraconazole, or with thenew oral triazole, saperconazole. Treatment with this last antimycotic requires adose of 100 <strong>to</strong> 200 mg per day for a period of three-<strong>and</strong>-a-half months (Franco etal., 1992).Because of their occupation, farmers, gardeners, <strong>and</strong> floriculturists are the personsmost exposed <strong>to</strong> the infection.The Disease in Animals: The disease in horses <strong>and</strong> mules is similar <strong>to</strong> that inhumans; it must be differentiated from epizootic lymphangitis caused by


354 MYCOSESHis<strong>to</strong>plasma farciminosum (Cryp<strong>to</strong>coccus farciminosum). The skin covering thespherical nodules becomes moist, the hair falls out, <strong>and</strong> a scab forms. The ulcersheal slowly <strong>and</strong> leave alopecic scars. The affected extremity swells due <strong>to</strong> lymphaticstasis. No cases of dissemination have been described in horses.The disease in dogs may manifest as the cutaneo-lymphatic form, but it frequentlyaffects the bones, liver, <strong>and</strong> lungs.The disease in cats is of particular interest because it has often served as thesource of infection for humans. One of these epizootic episodes occurred inMalaysia, where four veterinary students became infected when caring for cats withsporotrichosis on their forelegs <strong>and</strong> faces. Five cats with lesions inflicted duringfights in the clinic of the Veterinary School were treated with antibacterial medicationsfor two weeks, but the wounds did not heal. During this period, various ulcerativenodules appeared on the eyes, behind the ears, <strong>and</strong> in the nose. S. schenckii wasisolated from these lesions. The four students who treated the cats contractedsporotrichosis, as did the owner of one of the cats (Zamri-Saad et al., 1990). Threemembers of a family caught the infection from their cat <strong>and</strong> became ill with cutaneoussporotrichosis, which disappeared completely after two weeks of treatmentwith ke<strong>to</strong>conazole (Haqvi et al., 1993). Other cases of zoonotic transmissionoccurred in Brazil (Larsson et al., 1989) <strong>and</strong> the United States (Dunstan et al.,1986). Reed et al. (1993) described the case of a veterinarian who contracted theinfection from a cat; the authors also reviewed the relevant literature.Source of Infection <strong>and</strong> Mode of Transmission: The reservoirs of the fungus aresoil <strong>and</strong> plants. Humans <strong>and</strong> animals almost always become infected through a cutaneouslesion. The infection can be contracted from h<strong>and</strong>ling moss, wood splinters,firewood, or dead vegetation on which the fungus has developed. The source ofinfection in a gold mine epidemic in the Transvaal (South Africa) was timber onwhich S. schenckii was growing. Nevertheless, the source of infection is not alwayseasily recognized. Out of the 53 cases of sporotrichosis that occurred in the AyerzaLagoon area of Guatemala, 24 (45.3%) patients attributed the wound <strong>and</strong> subsequentulceration <strong>to</strong> h<strong>and</strong>ling fish, 6 (11.3%) attributed it <strong>to</strong> wood splinters, <strong>and</strong> 20 (37.7%)could not remember any trauma. An attempt <strong>to</strong> isolate S. schenckii from 58 environmentalsamples yielded negative results (Mayorga et al., 1979).Inhalation provides another entry route for the fungus <strong>and</strong> is responsible for thesmall number of pulmonary sporotrichosis cases that have been recorded.Feline sporotrichosis is known for its ability <strong>to</strong> transmit the infection <strong>to</strong> humans.Of 19 people who contracted the disease from a cat in the United States, none hadexperienced any traumatic lesion at the site of infection. Transmission occurredthrough direct contact with the ulcerous lesions on the cats’ skin, which contained alarge amount of fungus. The principal victims of zoonotic sporotrichosis are veterinarians.Of the 19 zoonotic cases, 12 involved veterinarians or their assistants(Dunstan et al., 1986). Outside the United States, transmission was attributed <strong>to</strong> catscratches or bites.Cats (usually male) may carry decaying vegetation containing the fungus betweentheir nails <strong>and</strong> may transmit the infection <strong>to</strong> other cats when they fight.Role of Animals in the Epidemiology of the Disease: Sporotrichosis is a disease<strong>common</strong> <strong>to</strong> man <strong>and</strong> animals. Feline sporotrichosis is zoonotic.


SPOROTRICHOSIS 355Diagnosis: Diagnosis can be confirmed by culture <strong>and</strong> identification of the fungus.A specific <strong>and</strong> rapid method is direct immunofluorescence applied <strong>to</strong> biopsysamples from affected tissues or smears from sputum <strong>and</strong> bronchial lavages.Serological tests (latex agglutination, immunodiffusion, indirect immunofluorescence)are useful for patients with extracutaneous sporotrichosis. The disadvantageof serological tests is that antibodies may take some time <strong>to</strong> develop or may disappearafter a while even though the disease persists (Pluss <strong>and</strong> Opal, 1986).Control: It is recommended that wood in industries where cases occur be treatedwith fungicides. Moss must be wetted only immediately prior <strong>to</strong> packing plants soas <strong>to</strong> keep the fungus from developing.Veterinarians <strong>and</strong> their assistants should use gloves <strong>to</strong> h<strong>and</strong>le <strong>and</strong> treat cats withcutaneous legions suspected of being sporotrichosis.BibliographyAinsworth, G.C., P.K.C. Austwick. Fungal Diseases of Animals. 2nd ed. Farnham Royal,Slough, United Kingdom: Commonwealth Agricultural Bureau; 1973.Benenson, A.S., ed. Control of Communicable Diseases in Man. 15th ed. An official repor<strong>to</strong>f the American Public Health Association. Washing<strong>to</strong>n, D.C.: American Public HealthAssociation; 1990.Bruner, D.W., J.H. Gillespie. Hagan’s Infectious Diseases of Domestic Animals. 6th ed.Ithaca, New York: Coms<strong>to</strong>ck; 1973.Coles, F.B., A. Schuchat, J.R. Hibbs, et al. A multistate outbreak of sporotrichosis associatedwith sphagnum moss. Am J Epidemiol 136:475–487, 1992.Dunstan, R.W., K.A. Reimann, R.F. Langham. Feline sporotrichosis. Zoonosis update. JAm Vet Med Assoc 189:880–883, 1986.Franco, L., I. Gómez, A. Restrepo. Saperconazole in the treatment of systemic <strong>and</strong> subcutaneousmycoses. Int J Derma<strong>to</strong>l 31:725–729, 1992.Haqvi, S.H., P. Becherer, S. Gudipati. Ke<strong>to</strong>conazole treatment of a family with zoonoticsporotrichosis. Sc<strong>and</strong> J Infect Dis 25:543–545, 1993.Larsson, C.E., M.A. Goncalves, V.C. Araujo, et al. Esporotricosis felina: aspec<strong>to</strong>s clínicose zoonóticos. Rev Inst Med Trop Sao Paulo 31:351–358, 1989.Lima, L.B., A.C. Pereira, Jr. Esprotricose-Inqueri<strong>to</strong> epidemiológico. Importancia comodoença profissional. An Bras Derma<strong>to</strong>l 56:243–248, 1981.Lurie, H.I. Five unusual cases of sporotrichosis from South Africa showing lesions in muscles,bones, <strong>and</strong> viscera. Br J Surg 50:585–591, 1962.Mackinnon, J.E. Ecology <strong>and</strong> epidemiology of sporotrichosis. In: Pan American HealthOrganization. Proceedings: International Symposium on Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>;1970. (Scientific Publication 205).Mayorga, R., A. Cáceres, C. Toriello, G. Gutiérrez, O. Álvarez, M.E. Ramírez, et al.Investigación de una zona endémica de esporotricosis en la región de la laguna de Ayarza,Guatemala. Bol Oficina Sanit Panam 87:20–34, 1979.Mitchell, T.G. Micosis subcutáneas. In: Joklik, W.K., H.P. Willet, D.B. Amos, eds. ZinsserMicrobiología. 17.ª ed. Buenos Aires: Edi<strong>to</strong>rial Médica Panamericana; 1983.Negroni, B. Micosis cutáneas y viscerales. 5.ª ed. Buenos Aires; López; 1972.Pluss, J.L., S.M. Opal. Pulmonary sporotrichosis: Review of treatment <strong>and</strong> outcome.Medicine 65:143–153, 1986.Reed, K.D., F.M. Moore, G.E. Geiger, M.E. Stemper. Zoonotic transmission of sporotrichosis:Case report <strong>and</strong> review. Clin Infect Dis 16:384–387, 1993.


356 MYCOSESRichard, J.L. Sporotrichosis. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger,eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.Zamri-Saad, M., T.S. Salmiyah, S. Jasni, et al. Feline sporotrichosis: An increasinglyimportant zoonotic disease in Malaysia. Vet Rec 127:480, 1990.ZYGOMYCOSISICD-10 B46.0 pulmonary mucormycosis; B46.1 rhinocerebral mucormycosis;B46.2 gastrointestinal mucormycosis; B46.3 cutaneous mucormycosis;B46.4 disseminated mucormycosis; B46.8 other zygomycosesSynonyms: Mucormycosis, en<strong>to</strong>mophthoromycosis.Etiology: Zygomycosis denotes a group of <strong>diseases</strong> caused by several genera <strong>and</strong>species of fungi belonging <strong>to</strong> the class Zygomycetes, orders En<strong>to</strong>mophthorales <strong>and</strong>Mucorales. Consequently, the etiologic agents are numerous; the principal ones arementioned below in connection with the different <strong>diseases</strong> they cause, which can besubdivided in<strong>to</strong> en<strong>to</strong>mophthoromycoses <strong>and</strong> mucormycoses (CIOMS, 1982).All the zygomycetes develop as hyphae <strong>and</strong> appear in the environment as well asin tissue as filamen<strong>to</strong>us fungi. Sabouraud’s agar is an excellent culture medium forthese fungi, in which they develop at ambient temperature. The sporangiophores(specialized hyphae that support sporangia) contain many asexual spores (Carter <strong>and</strong>Chengappa, 1991).Geographic Distribution: Worldwide. Mucormycosis has no defined geographicdistribution. En<strong>to</strong>mophthoromycosis predominates in the tropics, particularly inAfrica <strong>and</strong> Asia.Occurrence in Man: It occurs sporadically, particularly in patients weakened byother <strong>diseases</strong>. However, in the 1970s there was an epidemic of cutaneous zygomycosisin the United States caused by contamination of elastic b<strong>and</strong>ages with the fungus.The clinical manifestation was cellulitis, caused by direct inoculation of the fungusthrough the b<strong>and</strong>ages. The infection was invasive in some patients <strong>and</strong> affectedmuscles <strong>and</strong> internal organs (Sugar, 1990). At present, the incidence of zygomycosisis increasing because of the longer survival of diabetics <strong>and</strong> the growing number ofimmunosuppressed patients. Despite its broad diffusion in nature <strong>and</strong> the likelihoodthat humans will come in<strong>to</strong> contact with spores, this is not a very frequent mycosis.It is possible that the incidence of mucormycoses is higher in the developed countries,given the higher survival rate of diabetics <strong>and</strong> the number of immunosuppressedpatients. In a hospital in Washing<strong>to</strong>n, D.C., 730 cases of mucormycoses were recordedbetween 1966 <strong>and</strong> 1988. Of 170 cases of en<strong>to</strong>mophthoromycosis caused byBasidiobolus hap<strong>to</strong>sporus described up <strong>to</strong> 1975, 112 occurred in Africa. To these casesshould be added 75 cases in Ug<strong>and</strong>a that became known later (Kelly et al., 1980). Thisdisease also occurs in Southeast Asia <strong>and</strong> Latin America. En<strong>to</strong>mophthoromycosis due<strong>to</strong> the genus Conidiobolus also occurs in the tropics <strong>and</strong> is more <strong>common</strong> among men(CIOMS, 1982).


ZYGOMYCOSIS 357Occurrence in Animals: It occurs sporadically in many animal species, such asdomestic <strong>and</strong> wild mammals (including marine mammals), birds, reptiles, amphibians,<strong>and</strong> fish. There was a significant epizootic outbreak in New South Wales <strong>and</strong>Queensl<strong>and</strong>, Australia, affecting 52 sheep farms; 700 sheep died in three months. Thecausal agent was Conidiobolus incongruens of the order En<strong>to</strong>mophthorales (Carriganet al., 1992).The Disease in Man: The agents of mucormycoses are potential pathogens thatare classified as opportunistic, since they invade the tissues of patients debilitated byother <strong>diseases</strong> or treated for a long time with antibiotics or corticosteroids. About40% of the cases have been associated with diabetes mellitus. In contrast, in Africa<strong>and</strong> Asia en<strong>to</strong>mophthoromycoses occur in individuals without his<strong>to</strong>ries of preexistingillness (Bittencourt et al., 1982).The mucormycoses are caused by fungi of the genera Absidia, Mucor, Rhizopus,Cunninghamella, Rhizomucor, <strong>and</strong> several others. The infection begins in the nasalmucosa <strong>and</strong> paranasal sinuses, where the fungi may multiply rapidly <strong>and</strong> spread <strong>to</strong>the eye sockets, meninges, <strong>and</strong> brain. The clinical forms caused by these fungi arerhinocerebral, pulmonary, gastrointestinal, disseminated, cutaneous, <strong>and</strong> subcutaneousmucormycoses. The rhinocerebral form appears mainly in diabetes mellituspatients with acidosis <strong>and</strong> in leukemia patients with prolonged neutropenia. Patientshave fever, facial pain, <strong>and</strong> headache. As rhinocerebral mucormycosis progresses,there may be loss of vision, p<strong>to</strong>sis, <strong>and</strong> pupillary dilatation. This form of the diseaseis highly fatal. Patients with a malignant blood disease <strong>and</strong> those receiving immunosuppressantsprimarily suffer from pulmonary or disseminated mucormycoses <strong>and</strong>,less frequently, from the rhinocerebral form. The gastrointestinal form has occurredin a few cases in malnourished children <strong>and</strong> in adult patients with advanced malnutrition;it is generally diagnosed postmortem. The cutaneous <strong>and</strong> subcutaneous formmay be due <strong>to</strong> deep burns, injections, <strong>and</strong> application of contaminated b<strong>and</strong>ages.Mucormycosis is characterized by vascular occlusion with fungal hyphae, thrombosis,<strong>and</strong> necrosis.Localized mucormycosis may disseminate (disseminated mucormycosis) <strong>to</strong> variousorgans <strong>and</strong> systems. The underlying <strong>diseases</strong> are generally leukemia, solid neoplasias,chronic renal deficiency (dialysis treatment with deferoxamine seems <strong>to</strong>predispose the patient <strong>to</strong> mucormycosis, particularly <strong>to</strong> Rhisopus spp.), hepatic cirrhosis,organ transplants (particularly bone marrow transplants), <strong>and</strong> diabetes. Thelargest group of disseminated mucormycoses involves cancer patients (51% of 185cases analyzed) (Ingram et al., 1989).Treatment consists of controlling the underlying disease, controlling hyperglycemia<strong>and</strong> acidosis in diabetics, <strong>and</strong> reducing immunosuppressant use in othercases. Surgical intervention <strong>and</strong> systemic administration of amphotericin B yieldedfavorable results in pulmonary <strong>and</strong> rhinocerebral mucormycosis when diagnosisoccurred early. In primary cutaneous mucormycosis, débridement <strong>and</strong> <strong>to</strong>pical treatmentwith amphotericin B are indicated. Generally, the earlier the infection isdetected, the smaller the amount of dead tissue that will have <strong>to</strong> be removed <strong>and</strong> thegreater the chances for avoiding major tissue damage (Sugar, 1990).Treatment of en<strong>to</strong>mophthoromycosis consists primarily of surgical excision of thesubcutaneous nodules (Basidiobolus) or corrective surgery (Conidiobolus) of thenose <strong>and</strong> other parts of the face. It is advisable at the same time <strong>to</strong> treat the patient


358 MYCOSESwith ke<strong>to</strong>conazole or some other oral antimycotic azole derivative (Yangcoet al., 1984).En<strong>to</strong>mophthoromycoses due <strong>to</strong> Basidiobolus hap<strong>to</strong>sporus are characterized bythe formation of granulomas with eosinophilic infiltration in subcutaneous tissues.Generally, the region affected is the but<strong>to</strong>ck or thigh, with hard tumefaction of thesubcutaneous tissue <strong>and</strong> a clear delimitation from the healthy tissue. The disease isusually benign, but can sometimes be invasive <strong>and</strong> cause death (Greenham, 1979;Kelly et al., 1980).En<strong>to</strong>mophthoromycoses due <strong>to</strong> Conidiobolus coronatus <strong>and</strong> C. incongruens generallyoriginate in the lower nasal conchae <strong>and</strong> invade the subcutaneous facial tissues<strong>and</strong> paranasal sinuses. Lesions in the pericardium, mediastinum, <strong>and</strong> the lungshave also been described (CIOMS, 1982).The Disease in Animals: Zygomycosis in animals is usually found duringnecropsy or postmortem inspection in abat<strong>to</strong>irs. Few cases are confirmed by isolation<strong>and</strong> identification of the causal agent. Lesions are granuloma<strong>to</strong>us or ulcerative.Zygomycosis in cattle, sheep, <strong>and</strong> goats usually appears as ulcers of the abomasum.A 10-year study of gastrointestinal mycoses in cattle was conducted in Japan. Of 692cattle au<strong>to</strong>psied, 45 had systemic mycosis, 38 of them in the gastrointestinal tract.The large majority (94.7%) of s<strong>to</strong>mach infections were due <strong>to</strong> mucormycoses <strong>and</strong> thelesions consisted of focal hemorrhagic necroses. Many of the cattle were affected bypredisposing fac<strong>to</strong>rs for ruminal acidosis, such as ruminal a<strong>to</strong>ny (Chihaya et al.,1992). In cattle, lesions can also be found in nasal cavities <strong>and</strong> bronchial, mesenteric,<strong>and</strong> mediastinal nodes (Carter <strong>and</strong> Chengappa, 1991). In some countries, these fungiare an important cause of mycotic abortions. In Great Britain, they account for 32%of abortions caused by fungi, <strong>and</strong> in New Zeal<strong>and</strong> for 75%.In horses, zygomycosis takes the form of a chronic, localized disease that causesthe formation of cutaneous granulomas on the extremities. A clinical study of 266cases of zygomycosis conducted in tropical Australia found that 18% involvedBasidiobolus hap<strong>to</strong>sporus <strong>and</strong> 5.3% involved Conidiobolus coronatus.In the disease caused by B. hap<strong>to</strong>sporus (B. ranarum), lesions are found primarilyon the trunk <strong>and</strong> face. In contrast, lesions due <strong>to</strong> C. coronatus are located in thenasal region (Miller <strong>and</strong> Campbell, 1982). Pulmonary infection, disease of the gutturalpouch, systemic infection, <strong>and</strong> some mycotic abortions have also beendescribed in horses.Zygomycosis in piglets produces a gastric ulceration <strong>and</strong> appears in adult animalsas a disseminated infection. Gastroenteritis with diarrhea, dehydration, <strong>and</strong> somedeaths attributed <strong>to</strong> zygomycosis have been described in suckling pigs (Reed et al.,1987). Disseminated zygomycosis appears as granulomas in the submaxillary, cervical,<strong>and</strong> mesenteric nodes, <strong>and</strong> in the abdominopelvic organs. Three herd animalsweighing between 50 <strong>and</strong> 80 kg were found with very swollen subm<strong>and</strong>ibularnodes; systemic dissemination was confirmed postmortem in three of them (Sanfordet al., 1985).An epidemic occurred in 52 sheep farms in Australia, leading <strong>to</strong> the death of 700sheep within a period of three months. The affected animals had marked, asymmetricalswelling of the face, extending from the nostrils <strong>to</strong> the eyes. They weredepressed, without appetite, <strong>and</strong> had marked dyspnea <strong>and</strong> frequent bloody dischargefrom the nose. The animals would die between 7 <strong>and</strong> 10 days later. Necropsy con-


ZYGOMYCOSIS 359firmed severe necrogranuloma<strong>to</strong>us rhinitis that went as deep as the palate. Lesionswere also confirmed in the lymph nodes <strong>and</strong> thorax. Conidiobolus incongruens wasisolated from the nasal lesions, parotid gl<strong>and</strong>, subm<strong>and</strong>ibular gl<strong>and</strong>s, <strong>and</strong> the lungs.The most important his<strong>to</strong>pathological change was a severe granuloma<strong>to</strong>us inflammationthat contained small eosinophilic foci of coagulative necrosis. There werefungal hyphae in the center of these foci.To explain an outbreak of this magnitude, the authors assume that the infectionwas influenced by environmental fac<strong>to</strong>rs. After a rainy winter, grass grew plentifully;it was cut, <strong>and</strong> the cuttings began <strong>to</strong> decompose. Additional rain, heat, humidity,<strong>and</strong> the presence of decomposing plants created conditions favorable <strong>to</strong> proliferationof the etiologic agent (Carrigan et al., 1992).In dogs <strong>and</strong> cats, the disease usually affects the gastrointestinal tract <strong>and</strong> mortalityis very high. Lesions of the s<strong>to</strong>mach or small intestine are accompanied by vomiting,<strong>and</strong> lesions in the colon are accompanied by diarrhea <strong>and</strong> tenesmus (Ader, 1979).Source of Infection <strong>and</strong> Mode of Transmission: Zygomycetes are ubiqui<strong>to</strong>ussaprophytes that produce a large number of spores; they are <strong>common</strong> inhabitants ofdecomposing organic material <strong>and</strong> food, <strong>and</strong> are found in the gastrointestinal tract ofreptiles <strong>and</strong> amphibians. Humans contract the infection through inhalation, inoculation,<strong>and</strong> contamination of the skin by spores, <strong>and</strong> sometimes through ingestion. The<strong>common</strong> route of entry is the nose, by inhalation of spores. Debilitating <strong>diseases</strong>,such as diabetes mellitus, <strong>and</strong> prolonged treatment with immunosuppressants <strong>and</strong>antibiotics, are important causal fac<strong>to</strong>rs of mucormycosis. Mucoraceae spores probablydo not germinate in individuals with intact immune systems, judging fromexperimental tests in labora<strong>to</strong>ry animals. However, some cases have been describedin apparently normal people with no known underlying disease. Subcutaneous en<strong>to</strong>mophthoromycosisdue <strong>to</strong> Basidiobolus develops as a result of direct inoculation bythorns, <strong>and</strong> the disease caused by Conidiobolus spp. is contracted through inhalation.En<strong>to</strong>mophthoromycosis generally occurs in healthy individuals with no preexistingdisease.In domestic animals, the digestive route of infection seems <strong>to</strong> be more importantthan inhalation.Role of Animals in the Epidemiology of the Disease: Humans <strong>and</strong> animals contractthe infection from a <strong>common</strong> source in the environment. The infection is nottransmitted from one individual <strong>to</strong> another (man or animal).Diagnosis: Diagnosis is based on confirmation of the agent’s presence in scrapingsor biopsies of lesions by means of direct microscopic examination or by culture.Zygomycetes in tissue can be identified by their large nonseptate hyphae. The speciesof fungus can only be determined by culture <strong>and</strong> spore identification (Ader, 1979). Anindirect ELISA test with a homogenate of Rhizopus arrhizus <strong>and</strong> Rhizomucor pusilluscan be useful for diagnosing mucormycosis. This test was able <strong>to</strong> detect 33 of 43 casesof mucormycosis. The sensitivity of the test is 81% <strong>and</strong> the specificity is 94%. It cannotdetermine the genus or the species of the causal agent (Kaufman et al., 1989).Control: Human zygomycosis can be prevented in many cases by proper treatmen<strong>to</strong>f metabolic disorders, especially diabetes mellitus. Prolonged treatment withantibiotics <strong>and</strong> corticosteroids should be limited <strong>to</strong> those cases in which it isabsolutely necessary. Animals should not be allowed <strong>to</strong> consume moldy fodder.


360 MYCOSESBibliographyAder, P.L. Phycomycosis in fifteen dogs <strong>and</strong> two cats. J Am Vet Med Assoc 174:1216–1223, 1979.Ainsworth, G.C., P.K.C. Austwick. Fungal Diseases of Animals. 2nd ed. Farnham Royal,Slough, United Kingdom: Commonwealth Agricultural Bureau; 1973.Ajello, L., L.K. Georg, W. Kaplan, L. Kaufman. Labora<strong>to</strong>ry Manual for MedicalMycology. Washing<strong>to</strong>n, D.C.: U.S. Government Printing Office; 1963. (Public HealthService Publication 994).Bittencourt, A.L., G. Serra, M. Sadigursky, M.G.S. Araujo, M.C.S. Campos, L.C.M.Sampaio. Subcutaneous zygomycosis caused by Basidiobolus hapthoporus: Presentation of acase mimicking Burkitt’s lymphoma. Am J Trop Med Hyg 31:370–373, 1982.Carter, G.R. Diagnostic Procedures in Veterinary Microbiology. 2nd ed. Springfield:Thomas; 1973.Carter, G.R., M.M. Chengappa. Essentials of Veterinary Bacteriology <strong>and</strong> Mycology. 4thed. Philadelphia: Lea & Febiger; 1991.Carrigan, M.J., A.C. Small, G.H. Perry. Ovine nasal zygomycosis caused by Conidiobolusincongruus. Aust Vet J 69:237–240, 1992.Chihaya, Y., K. Matsukawa, K. Ohshima, et al. A pathological study of bovine alimentarymycosis. J Comp Pathol 197:195–206, 1992.Council for International Organizations of Medical Sciences (CIOMS). Vol 2: InfectiousDiseases; Part 2: Mycoses. In: International Nomenclature of Diseases. Geneva:CIOMS; 1982.González-Mendoza, A. Opportunistic mycoses. In: Pan American Health Organization.Proceedings: International Symposium on Mycoses. Washing<strong>to</strong>n, D.C.: <strong>PAHO</strong>; 1970.(Scientific Publication 205).Greenham, R. Subcutaneous phycomycosis: Not always benign. Lancet 1:97:98, 1979.Ingram, C.W., J. Sennesh, J.N. Cooper, J.R. Perfect. Disseminated zygomycosis: Report offour cases <strong>and</strong> review. Rev Infect Dis 11:741–754, 1989.Kaufman, L., L.F. Turner, D.W. McLaughlin. Indirect enzyme-linked immunosorbent assayfor zygomycosis. J Clin Microbiol 27:1979–1982, 1989.Kelly, S., N. Gill, M.S.R. Hutt. Subcutaneous phycomycosis in Sierra Leone. Trans Roy SocTrop Med Hyg 74:396–397, 1980.Miller, R.I., R.S.F. Campbell. Clinical observations on equine phycomycosis. Aust Vet J58:221–226, 1982.Reed, W.M., C. Hanika, N.A.Q. Mehdi, C. Shackelford. Gastrointestinal zygomycosis insuckling pigs. J Am Vet Med Assoc 191:549–550, 1987.Richard, J.L. Phycomycoses. In: Hubbert, W.T., W.F. McCulloch, P.R. Schnurrenberger,eds. Diseases Transmitted from Animals <strong>to</strong> Man. 6th ed. Springfield: Thomas; 1975.Sanford, S.E., G.K.A. Josephson, E.H. Waters. Subm<strong>and</strong>ibulary <strong>and</strong> disseminated zygomycosis(mucormycosis) in feeder pigs. J Am Vet Med Assoc 186:171–174, 1985.Soltys, M.A. Bacteria <strong>and</strong> Fungi Pathogenic <strong>to</strong> Man <strong>and</strong> Animals. London: Baillière-Tindall; 1963.Sugar, A.M. Agents of mucormycosis <strong>and</strong> related species. In: M<strong>and</strong>ell, G.L., R.G. Douglas,Jr., J.E. Bennett, eds. Principles <strong>and</strong> Practice of Infectious Diseases. 3rd ed. New York:Churchill Livings<strong>to</strong>ne Inc.; 1990.Yangco, B.G., J.I. Okafor, D. TeStrake. In vitro susceptibilities of human <strong>and</strong> wild-typeisolates of Basidiobolus <strong>and</strong> Conidiobolus species. Antimicrob Agents Chemother 25:413–416, 1984.


INDEXAAbortionbrucellosis, 43, 44-49, 55, 61, 62campylobacteriosis, 69, 72, 73-75, 76colibacillosis, 94, 95contagious (see Brucellosis)epizooticsheep (see Diseases caused byCampylobacter fetus)vibrionic (see Diseases caused byCampylobacter fetus)(see also Brucellosis)infectious (see Brucellosis)lep<strong>to</strong>spirosis, 159, 160, 161listeriosis, 170-173, 175, 176nocardiosis, 196salmonellosis, 238strep<strong>to</strong>coccosis, 260tetanus, 266, 268, 269tularemia, 278yersiniosisenterocolitic, 126pseudotuberculous, 220Absidia, 357Acremonium, 345Actinobacillus, 142, 199Actinomaduramadurae, 345, 346pelletieri, 345Actinomyces, 3, 5bovis, 3-5israelii, 3-5meyeri, 3naeslundi, 3odon<strong>to</strong>lytical, 3suis, 4viscosus, 3, 4Actinomycetales, 103, 195, 229Actinomyce<strong>to</strong>ma (see Myce<strong>to</strong>ma)Actinomycosis, 3-6Actinostrep<strong>to</strong>trichosis, (seeActinomycosis)Adiaspiromycosis, 303-305Adiaspirosis (see Adiaspiromycosis)Aedes aegypti, 188Aeromonas, 6-8, 10-12caviae, 6, 8hydrophila, 6-12j<strong>and</strong>ae, 6salmonicida, 6, 7schuberti, 6sobria, 6-9, 11, 12trota, 6, 9veronii, 6Aeromoniasis, 6-14Afipia felis, 78, 80Allantiasis (see Botulism)Allescheria boydii (seePseudallescheria boydii)Alliga<strong>to</strong>rs, animal erysipelas, 14Alpacasbrucellosis, 49listeriosis, 171tuberculosis, zoonotic, 292Amblyomma americanum, 279Amphibiansaeromoniasis, 8, 9, 10, 12salmonellosis, 236zygomycosis, 357, 359Animals, domesticaeromoniasis, 11anthrax, 21, 25aspergillosis, 305, 306, 308botulism, 33, 35brucellosis, 41, 49, 50campylobacteriosis, 68, 69-70coccidioidomycosis, 321cryp<strong>to</strong>coccosis, 327, 328derma<strong>to</strong>philosis, 104, 105derma<strong>to</strong>phy<strong>to</strong>sis, 335<strong>diseases</strong> caused by nontuberculousmycobacteria, 109food poisoningclostridial, 82staphylococcal, 252, 254his<strong>to</strong>plasmosis, 340, 342lep<strong>to</strong>spirosis, 158, 162-165listeriosis, 171, 172-173Lyme disease, 180, 181melioidosis, 185, 186myce<strong>to</strong>ma, 346nocardiosis, 195pasteurellosis, 199, 203pro<strong>to</strong>thecosis, 348-349rhinosporidiosis, 351rhodococcosis, 229salmonellosis, 236, 237, 239, 241tetanus, 268tuberculosis, zoonotic, 291, 294361


362 INDEXtularemia, 276, 279, 280Vibrio cholerae, non-O1, 118yersiniosis, pseudotuberculous, 219,223zygomycosis, 357-359(see also individual species)Animals, labora<strong>to</strong>ryaeromoniasis, 10anthrax, 23, 26colibacillosis, 96corynebacteriosis, 100melioidosis, 186, 188plague, 215rat-bite fever, 227tuberculosis, zoonotic, 291tularemia, 280(see also individual species)Animals, wildadiaspiromycosis, 303anthrax, 23, 24, 25, 26aspergillosis, 305, 306, 308brucellosis, 49-50campylobacteriosis, 68, 70coccidioidomycosis, 321corynebacteriosis, 100cryp<strong>to</strong>coccosis, 328derma<strong>to</strong>philosis, 104, 105his<strong>to</strong>plasmosis, 340, 342leprosy, 153lep<strong>to</strong>spirosis, 158, 161, 163listeriosis, 171, 173Lyme disease, 180-182melioidosis, 186nocardiosis, 195pasteurellosis, 199, 203rat-bite fever, 227relapsing fever, tick-borne, 272salmonellosis, 236, 240sporotrichosis, 353tuberculosis, zoonotic, 291, 293tularemia, 276, 278, 280yersiniosisenterocolitic, 124, 125pseudotuberculous, 219, 221, 223zygomycosis, 357(see also individual species)Animals, zooaeromoniasis, 10anthrax, 24blas<strong>to</strong>mycosis, 312c<strong>and</strong>idiasis, 317cryp<strong>to</strong>coccosis, 328<strong>diseases</strong> caused by nontuberculousmycobacteria, 112, 113gl<strong>and</strong>ers, 144leprosy, 150melioidosis, 185salmonellosis, 240shigellosis, 248, 249, 250tuberculosis, 107, 109, 111zoonotic, 291-292tularemia, 280yersiniosis, pseudotuberculous, 221(see also individual species)Antelopes, 50Anthrax, 21-28bacterial (see Anthrax)hematic (see Anthrax)transmission cycle, figure, 25Apodemus sylvaticus, 181Arachnia propionica, 3Argusminiatus, 272persicus, 272Arizona hinshawii, 240Armadilloscat-scratch disease, 78<strong>diseases</strong> caused by nontuberculousmycobacteria, 113leprosy, 150-154relapsing fever, tick-borne, 272Arthritis, Lyme (see Lyme disease)Arthropodsbrucellosis, 50derma<strong>to</strong>philosis, 105leprosy, 153plague, 214tularemia, 276, 277, 279, 281Arvicanthis niloticus, 49Aspergillosis, 305-310Aspergillus, 305-309flavus, 305, 308fumigatus, 305, 307-309nidulans, 305niger, 305, 307parasiticus, 305terreus, 305, 308Baboons (see Monkeys; see Primates)Bacillusanthracis, 21, 23-27cereus, 21B


INDEX 363whitmori (see Yersinia enterocolitica)Bacterioidaceae, 190Bacterioses, 3-299Bacterium enterocoliticum (see Yersiniaenterocolitica)Bacteroides, 190-192asaccharolyticus, 192fragilis, 191, 192meleninogenicus, 192nodosus, 191-194Badgers, tuberculosis, 292Bang’s disease (see Brucellosis)Bar<strong>to</strong>nella henselae, 79Basidiobolushap<strong>to</strong>sporus, 356, 358ranarum, 358Batshis<strong>to</strong>plasmosis, 342, 343relapsing fever, tick-borne, 272shigellosis, 249Beavers, 276, 279Birdsadiaspiromycosis, 304aeromoniasis, 9, 11anthrax, 25aspergillosis, 305, 308, 309botulism, 28, 32, 33, 36-38brucellosis, 50campylobacteriosis, 68-71c<strong>and</strong>idiasis, 316, 317, 319colibacillosis, 92, 96cryp<strong>to</strong>coccosis, 328derma<strong>to</strong>phy<strong>to</strong>sis, 336<strong>diseases</strong> caused by nontuberculousmycobacteria, 107, 109, 111, 113-115erysipelas, animal, 14, 15, 17food poisoningclostridial, 83, 84staphylococcal, 252, 254his<strong>to</strong>plasmosis, 342infection of wounds, clostridial, 88listeriosis, 171, 173, 175Lyme disease, 182pasteurellosis, 199, 200, 203relapsing fever, tick-borne, 272salmonellosis, 233, 236, 239-240,242-244sporotrichosis, 353tuberculosisavian, 113zoonotic, 285, 288, 292, 293tularemia, 277Vibrio cholerae, non-O1, 118yersiniosisenterocolitic, 124pseudotuberculous, 219, 221-223zygomycosis, 357Bison, Americanbrucellosis, 50pasteurellosis, 201Bison bison, 50, 119Black death (see Plague)Blas<strong>to</strong>myces dermatitidis, 311Blas<strong>to</strong>mycosis, 311-315European (see Cryp<strong>to</strong>coccosis)North American (see Blas<strong>to</strong>mycosis)Bordetella bronchiseptica, 202-203Borrelia, 179, 271-273anserina, 272brasiliensis, 271burgdorferi, 179, 180, 181-182caucasica, 271dut<strong>to</strong>ni, 271, 273hermsii, 271hispanica, 271parkeri, 271recurrentis, 271theileri, 272turicata, 271venezuelensi, 271Borreliosis (see Tick-borne relapsingfever)Lyme (see Lyme disease)Bos grunniens, 49Botulism, 28-40cases <strong>and</strong> deaths reported, USA,figure, 30cases reported, Argentina, figure, 32foods involved, USA, table, 31Bovinesactinomycosis, 4aeromoniasis, 11anthrax, 24, 26, 27aspergillosis, 305, 307, 308botulism, 28, 32, 33, 35-38brucellosis, 40, 41, 42, 44-47, 48, 49,50, 51, 52-53, 56-58, 60, 62campylobacteriosis, 69, 70, 72, 73-76c<strong>and</strong>idiasis, 317coccidioidomycosis, 321, 322colibacillosis, 91, 92, 95, 96, 97corynebacteriosis, 100, 101, 102cryp<strong>to</strong>coccosis, 327, 328


364 INDEXderma<strong>to</strong>philosis, 104, 105derma<strong>to</strong>phy<strong>to</strong>sis, 332, 335-337<strong>diseases</strong> caused by nontuberculousmycobacteria, 107, 110, 111, 114enterocolitis due <strong>to</strong> Clostridiumdifficile, 133erysipelas, animal, 16food poisoningclostridial, 83-85staphylococcal, 252his<strong>to</strong>plasmosis, 340, 342infectioncaused by Capnocy<strong>to</strong>phagacanimorsus, 147caused by Capnocy<strong>to</strong>phagacynodegmi, 147of wounds, clostridial, 88infertility, epizootic (see Diseasescaused by Campylobacter fetus)lep<strong>to</strong>spirosis, 158, 159-160, 162,165-166listeriosis, 171, 172-173, 175Lyme disease, 180, 181melioidosis, 185, 186, 187necrobacillosis, 192-193nocardiosis, 195, 196, 198pasteurellosis, 199, 201, 203, 204pro<strong>to</strong>thecosis, 348, 349relapsing fever, tick-borne, 272respira<strong>to</strong>ry disease complex (seePasteurellosis)rhinosporidiosis, 351rhodococcosis, 231salmonellosis, 236, 237, 240, 243sporotrichosis, 353strep<strong>to</strong>coccosis, 258, 260, 261tetanus, 266, 268tuberculosis, zoonotic, 283, 284, 285,286-288, 289, 290, 291-296Vibrio cholerae, non-O1, 118yersiniosisenterocolitic, 126pseudotuberculous, 220, 222zygomycosis, 358Bronchomycosis (see Aspergillosis)Brucella, 40-62, 72, 129abortus, 40-41, 43-45, 47-50, 52, 53,55, 56, 60, 62canis, 40, 43, 49, 50, 55, 56, 59, 62melitensis, 40-45, 47-52, 53, 56, 59,61-62neo<strong>to</strong>mae, 40, 43, 49ovis, 40, 43, 48, 50, 53, 54, 56, 59, 62suis, 40-43, 47-50, 51, 56, 62Brucellosis, 40-67abortion, 40, 43-49, 55, 61, 62bovine, transmission, mode of,figure, 52caprine, transmission, mode of,figure, 54contagious, 40, 43-49, 55, 61, 62epizootic, 40, 43-49, 55, 61, 62infectious, 40, 43-49, 55, 61, 62ovine, transmission, mode of,figure, 54swine, transmission, mode of,figure, 54Bubalus bubalis, 49, 112Buffaloesanthrax, 22, 23brucellosis, 41, 45, 49corynebacteriosis, 101<strong>diseases</strong> caused by nontuberculousmycobacteria, 112lep<strong>to</strong>spirosis, 163pasteurellosis, 201, 204tuberculosis, zoonotic, 293yersiniosis, enterocolitic, 126Bulls (see Bovines)Busse-Buschke’s disease (seeCryp<strong>to</strong>coccosis)CCamelsbrucellosis, 41, 49corynebacteriosis, 100, 101enterocolitis due <strong>to</strong> Clostridiumdifficile, 133pasteurellosis, 201plague, 211, 213sporotrichosis, 353Camelus bactrianus, 49Camelus dromedarius, 49Campylobacter, 67-76, 124coli, 67, 70fetus, 67, 69, 72-76var. intestinalis, 67, 69, 72, 73var. venerealis, 67, 72, 73-74, 76jejuni, 67-72, 73, 74laridis, 67upsaliensis, 67, 69Campylobacteriosis, 67-78disease caused by Campylobacterfetus, 72-78


INDEX 365transmission, probable mode of,figure, 75enteritis caused by Campylobacterjejuni, 67-72transmission, mode of, figure, 70Canaries (see Birds)Cancer, m<strong>and</strong>ibles (see Actinomycosis)C<strong>and</strong>ida, 315-318albicans, 315, 317, 318guillermondi, 315krusei, 315lusitaniae, 315parapsilosis, 315pseudotropicalis, 315tropicalis, 315C<strong>and</strong>idiasis, 315-320C<strong>and</strong>idomycosis (see C<strong>and</strong>idiasis)C<strong>and</strong>idosis (see C<strong>and</strong>idiasis)Capnocy<strong>to</strong>phagacanimorsus, 146-148cynodegmi, 146-148Caprinesanthrax, 21, 24, 26brucellosis, 42, 43, 47-48, 50, 53, 58,60, 61-62corynebacteriosis, 100, 101, 102cryp<strong>to</strong>coccosis, 327, 328derma<strong>to</strong>philosis, 104derma<strong>to</strong>phy<strong>to</strong>sis, 335-336food poisoning, clostridial, 83, 84infection of wounds, clostridial, 88lep<strong>to</strong>spirosis, 161listeriosis, 172melioidosis, 185, 186, 188rhodococcosis, 231salmonellosis, 238strep<strong>to</strong>coccosis, 260, 262tuberculosis, zoonotic, 289, 293, 294Vibrio cholerae, non-O1, 118yersiniosis, pseudotuberculous, 220zygomycosis, 358Carbuncle (see Anthrax)Caribou, 49, 51Carnivoresadiaspiromycosis, 304brucellosis, 50tuberculosis, zoonotic, 292yersiniosis, enterocolitic, 125Cas<strong>to</strong>r canadensis, 276, 279Cat-scratch disease, 78-81Cat-scratch syndrome (see Cat-scratchdisease)Catsadiaspiromycosis, 304blas<strong>to</strong>mycosis, 312brucellosis, 49campylobacteriosis, 69, 71c<strong>and</strong>idiasis, 317cat-scratch disease, 78-81cryp<strong>to</strong>coccosis, 327, 328, 330derma<strong>to</strong>philosis, 104derma<strong>to</strong>phy<strong>to</strong>sis, 332-337<strong>diseases</strong> caused by nontuberculousmycobacteria, 112enterocolitis due <strong>to</strong> Clostridiumdifficile, 133food poisoning, staphyloccocal, 252,255his<strong>to</strong>plasmosis, 342infection caused by Capnocy<strong>to</strong>phagacanimorsus, 146, 147, 148infection caused by Capnocy<strong>to</strong>phagacynodegmi, 146, 147, 148lep<strong>to</strong>spirosis, 161melioidosis, 186myce<strong>to</strong>ma, 346nocardiosis, 195, 197pasteurellosis, 200, 203plague, 208, 211, 213, 214pro<strong>to</strong>thecosis, 349rat-bite fever, 228relapsing fever, tick-bornerhodococcosis, 231salmonellosis, 236, 239sporotrichosis, 353-355tuberculosis, 112zoonotic, 285, 290-291, 293, 294tularemia, 276, 278, 279-280yersiniosisenterocolitic, 124, 125-127pseudotuberculous, 220zygomycosis, 359Cavern disease (see His<strong>to</strong>plasmosis)Cellulitis, anaerobic (see Clostridialwound infections)Cercocebus atys, 151Cervidae (see Deer)Chae<strong>to</strong>phractus villosus, 113Chicago disease (see Blas<strong>to</strong>mycosis)Chickensaspergillosis, 306, 308botulism, 33campylobacteriosis, 69, 70, 71c<strong>and</strong>idiasis, 316, 317


366 INDEXcolibacillosis, 96erysipelas, animal, 17listeriosis, 174relapsing fever, tick-borne, 272salmonellosis, 235, 239, 240, 242,244strep<strong>to</strong>coccosis, 260yersiniosis, enterocolitic, 128Chimpanzees, 150, 152Chinchillas, 125Chipmunks, Lyme disease, 180(see also Rodents)Chiropterans (see Bats)Cholera, 117-122fowl (see Pasteurellosis)Chrysosporium, 303-304crescens, 303, 304parvum, 303Citellus beecheyi, 212Clethrionomys, 181Clostridialfood poisoning, 82-87wound infections, 87-89Clostridium, 28, 29, 34, 82, 87, 88, 132,265argentinense, 29baratii, 29, 34botulinum, 28-29, 33, 34-37butyricum, 29, 34chauvoei, 89difficile, 132-136fallax, 87his<strong>to</strong>lyticum, 87, 88novyi, 87, 88perfringens, 82-86, 87, 88, 135septicum, 87, 88, 89sordellii, 87, 88, 135tetani, 265, 267, 268-269welchii (see C. perfringens)Coccidioides, 320immitis, 320-323Coccidioidomycosis, 320-325Cochliolobus spicifer, 345Colibacillosis, 90-99Colibacteriosis (see Colibacillosis)Coli<strong>to</strong>xemia (see Colibacillosis)Columba palumbus, 221, 223Conidiobolus, 356-359coronatus, 358incongruens, 357-359Corvus corvix, 50Corynebacteriosis, 93, 99-103Corynebacterium, 99-102bovis, 100, 101, 102cystitidis, 100, 101, 102diphtheriae, 99-100, 101kutscheri, 100, 101pilosum, 100, 101, 102pseudotuberculosis, 99-101, 102pyogenes, 191, 193renale, 99, 100, 101, 102ulcerans, 100, 101, 102Cows (see Bovines)Crocodiles, animal erysipelas, 14Crowsbrucellosis, 50Vibrio cholerae, non-O1, 118Crustaceanserysipelas, animal, 18poisoning caused by Vibrioparahaemolyticus, 140, 141Cryp<strong>to</strong>coccosis, 326-331Cryp<strong>to</strong>coccusfarciminosum (see His<strong>to</strong>plasmafarciminosum)neoformans, 326, 327, 328-330Cunninghamella, 357Curvularia geniculata, 345, 346Cynomys, 212, 213Cyprinus carpio, 9Cy<strong>to</strong>mycosis, reticuloendothelial (seeHis<strong>to</strong>plasmosis)DDarling’s disease (see His<strong>to</strong>plasmosis)Dasypushybridus, 113novemcinctus, 149, 150, 152Deercolibacillosis, 92corynebacteriosis, 101derma<strong>to</strong>philosis, 103Lyme disease, 180, 182rhodococcosis, 231tuberculosis, zoonotic, 291, 294yersiniosis, pseudotuberculous, 222Deer-fly fever (see Tularemia)Dermacen<strong>to</strong>r, 278, 279Dermatitis, mycotic (seeDerma<strong>to</strong>philosis)Derma<strong>to</strong>mycosis (see Derma<strong>to</strong>phy<strong>to</strong>sis)Derma<strong>to</strong>philosis, 103-106Derma<strong>to</strong>philuscongolensis, 103, 104, 105


INDEX 367derma<strong>to</strong>nomous (see D. congolensis)pedis (see D. congolensis)Derma<strong>to</strong>phy<strong>to</strong>sis, 332-339Diarrheaassociated with antibiotics (seeEnterocolitis due <strong>to</strong> Clostridiumdifficile)enteropathogenic (see Colibacillosis)Didelphisazarae, 50virginiana, 180Diphtheria, calf (see Necrobacillosis)DiseaseBang’s (see Brucellosis)bovine respira<strong>to</strong>ry disease complex(see Pasteurellosis)Busse-Buschke’s (seeCryp<strong>to</strong>coccosis)cat-scratch, 78-81cavern (see His<strong>to</strong>plasmosis)Chicago (see Blas<strong>to</strong>mycosis)Darling’s (see His<strong>to</strong>plasmosis)deer-fly (see Tularemia)Francis’ (see Tularemia)Gilchrist’s (see Blas<strong>to</strong>mycosis)Hansen’s (see Leprosy)Lyme, 179-184Ohara’s (see Tularemia)Posada’s (see Coccidioidomycosis)ray fungus (see Actinomycosis)red leg, 10Schmorl’s (see Necrobacillosis)Stuttgart (see Lep<strong>to</strong>spirosis)swineherd’s (see Lep<strong>to</strong>spirosis)Weil’s (see Lep<strong>to</strong>spirosis)Whitmore (see Melioidosis)Diseasescaused by Campylobacter fetus (seealso Campylobacteriosis), 72-78caused by nontuberculousmycobacteria, 107-117caused by non-O1 Vibrio cholerae,117-122Dogsactinomycosis, 3, 4anthrax, 24aspergillosis, 307blas<strong>to</strong>mycosis, 312, 313, 314botulism, 33, 37brucellosis, 43, 49, 50, 55, 59, 62campylobacteriosis, 69, 70, 71c<strong>and</strong>idiasis, 317coccidioidomycosis, 321, 323cryp<strong>to</strong>coccosis, 328derma<strong>to</strong>phy<strong>to</strong>sis, 332, 333, 334, 335,336<strong>diseases</strong> caused by nontuberculousmycobacteria, 112enterocolitis due <strong>to</strong> Clostridiumdifficile, 133, 135food poisoning, staphylococcal, 252,254gl<strong>and</strong>ers, 144his<strong>to</strong>plasmosis, 340, 342, 343infectioncaused by Capnocy<strong>to</strong>phagacanimorus, 146, 147, 148caused by Capnocy<strong>to</strong>phagacynodegmi, 146, 147, 148lep<strong>to</strong>spirosis, 158, 161, 162, 165listeriosis, 173Lyme disease, 180, 181, 182melioidosis, 185, 186myce<strong>to</strong>ma, 346nocardiosis, 195, 196-197pasteurellosis, 200, 203plague, 213, 214, 215, 216pro<strong>to</strong>thecosis, 348-349rhinosporidiosis, 351salmonellosis, 236, 239, 241shigellosis, 249sporotrichosis, 353, 354tetanus, 268tuberculosis, 112zoonotic, 285, 287, 290, 293, 294tularemia, 276Vibrio cholerae, non-O1, 118yersiniosisentercolitic, 123, 124, 125-128pseudotuberculous, 221, 223zygomycosis, 359Dolphinsblas<strong>to</strong>mycosis, 312erysipelas, animal, 14melioidosis, 185Doves (see Birds)Ducksbotulism, 33, 36, 37campylobacteriosis, 70<strong>diseases</strong> caused by nontuberculousmycobacteria, 113erysipelas, animal, 16relapsing fever, tick-borne, 272salmonellosis, 239, 240


368 INDEXtuberculosis, 113yersiniosis, pseudotuberculous, 219Dusicyongriseus, 50gymnocercus, 50Dysentery, bacillary (see Shigellosis)EEdema, malignant (see Infection,clostridial, of wounds)Edwardsiella, 240Ehrlichia ristici, 308Elephants, 23Emmonsia (see Chrysosporium)Emmonsiela capsulata, 339Endocarditis, 341Enteritiscaused by Campylobacter jejuni (seealso Campylobacteriosis), 67-72enterocolitis due <strong>to</strong> Clostridiumdifficile, 132-137vibrionic (see Enteritis caused byCampylobacter jejuni)Enterobacteriaceae, 233Enterocolitic yersiniosis, 122-132Enterocolitisdue <strong>to</strong> Clostridium difficile, 132-137hemorrhagic necrotizing (seeEnterocolitis due <strong>to</strong> Clostridiumdifficile)pseudomembranous (see Enterocolitisdue <strong>to</strong> Clostridium difficile)En<strong>to</strong>mophthorales, 356, 357En<strong>to</strong>mophthoromycosis (seeZygomycosis)Epidermophy<strong>to</strong>n floccosum, 332, 333,334Epididymitis, rams, 43, 48, 53, 59, 62Equinesactinomycosis, 4anthrax, 24, 26, 27aspergillosis, 307blas<strong>to</strong>mycosis, 312botulism, 33, 35brucellosis, 48c<strong>and</strong>idiasis, 317coccidioidomycosis, 321, 322, 323colibacillosis, 95corynebacteriosis, 100, 101, 102cryp<strong>to</strong>coccosis, 327, 328derma<strong>to</strong>philosis, 104derma<strong>to</strong>phy<strong>to</strong>sis, 335, 337enterocolitis due <strong>to</strong> Clostridiumdifficle, 133food poisoning, clostridial, 84, 85gl<strong>and</strong>ers, 143, 144his<strong>to</strong>plasmosis, 340, 342infection of wounds, clostridial, 88lep<strong>to</strong>spirosis, 160Lyme disease, 180, 181melioidosis, 185, 186, 187myce<strong>to</strong>ma, 346phthisis, nasal (see Gl<strong>and</strong>ers)rhinosporidiosis, 351rhodococcosis, 230, 231, 232salmonellosis, 238sporotrichosis, 353strep<strong>to</strong>coccosis, 258, 260tetanus, 266, 268, 269tuberculosis, zoonotic, 290, 294tularemia, 278zygomycosis, 358Erysipelas, animal <strong>and</strong> humanerysipeloid, 14-21transmission, mode of, figure, 17Erysipeloid, Rosenbach’s (seeErysipelas, animal <strong>and</strong> humanerysipeloid)Erysipelothrixrhusiopathiae, 14-18<strong>to</strong>nsillarum, 14Erysipelotrichosis (see Erysipelas,animal <strong>and</strong> human erysipeloid)Erythemachronicum migrans with polyarthritis(see Lyme disease)epidemic arthritic (see Rat-bite fever)migrans (see Erysipelas, animal <strong>and</strong>human erysipeloid)migrans with polyarthritis (see Lymedisease)Escherichia coli, 55, 90-97, 120, 247,250, 259enteroaggregative, 90, 94, 96enterohemorrhagic, 90-92, 94, 95, 96enteroinvasive, 90, 93-94, 96enteropathogenic, 90, 94, 96entero<strong>to</strong>xigenic, 90, 92-93, 94, 95,96, 97Eume<strong>to</strong>pias jubata, 312Eumycotic myce<strong>to</strong>ma (see Myce<strong>to</strong>ma)European blas<strong>to</strong>mycosis (seeCryp<strong>to</strong>coccosis)


INDEX 369Exophiala jeanselmei, 345FFarcy (see Gl<strong>and</strong>ers)Ferretsaeromoniasis, 11blas<strong>to</strong>mycosis, 312brucellosis, 50rat-bite fever, 228Fevercane-cutter’s (see Lep<strong>to</strong>spirosis)canicola (see Lep<strong>to</strong>spirosis)cat-scratch (see Cat-scratch disease)deer-fly (see Tularemia)desert (see Coccidioidomycosis)endemic relapsing (see Tick-bornerelapsing fever)Haverhill (see Rat-bite fever)Malta (see Brucellosis)Mediterranean (see Brucellosis)mud (see Lep<strong>to</strong>spirosis)pestilential (see Plague)rabbit (see Tularemia)rat-bite, 226-229rice-field (see Lep<strong>to</strong>spirosis)San Joaquin Valley (seeCoccidioidomycosis)shipping (see Pasteurellosis)spirillary (see Rat-bite fever)spirochetal (see Tick-borne relapsingfever)splenic (see Anthrax)strep<strong>to</strong>bacillary (see Rat-bite fever)swamp (see Lep<strong>to</strong>spirosis)tick-borne relapsing, 271-274Argus miniatus, 272Argus persicus, 272transmission, mode of, figure, 273transport (see Pasteurellosis)undulant (see Brucellosis)Fishaeromoniasis, 6-7, 8, 9-10, 11, 12<strong>diseases</strong> caused by nontuberculousmycobacteria, 110, 112, 113, 115erysipelas, animal, 18listeriosis, 171melioidosis, 185nocardiosis, 195pro<strong>to</strong>thecosis, 349Vibrio parahaemolyticus, 138, 139,140, 141zygomycosis, 357Fleasmelioidosis, transmitted by, 188plague, transmitted by, 207, 211, 213,214, 215tularemia, 276, 280Fliesderma<strong>to</strong>philosis, 105salmonellosis, 242shigellosis, 249Food poisoningcaused by Vibrio parahaemolyticus,138-142clostridial, 82-87staphylococcal, 251-257Foot-rot (see Necrobacillosis)Fowlbotulism, 33, 36campylobacteriosis, 69colibacillosis, 92derma<strong>to</strong>phy<strong>to</strong>sis, 336<strong>diseases</strong> caused by nontuberculousmycobacteria, 113tetanus, 268tuberculosis, 113Foxesbrucellosis, 50lep<strong>to</strong>spirosis, 158tuberculosis, zoonotic, 292tularemia, 276Francis’ disease (see Tularemia)Francisella tularensis, 275-278, 280-281Frogsaeromoniasis, 10disease caused by nontuberculousmycobacteria, 112(see also Poikilotherms)Fusobacterium necrophorum,190-193GGalictis furax-huronax, 50Gangrene, gas (see Clostridial woundinfection)Gastroenteritisaeromoniasis, 8-9anthrax, 23campylobacteriosis, 73clostridial (see Clostridial foodpoisoning)colibacillosis, 92


370 INDEXfood poisoning caused by Vibrioparahaemolyticus, 139lep<strong>to</strong>spirosis, 161melioidosis, 186rhodococcosis, 231salmonellosis, 234, 235, 236-237,242staphylococcal (see Staphylococcalfood poisoning)tularemia, 277Vibrio cholerae, non-O1, 118yersiniosisenterocolitic, 124, 127, 129pseudotuberculous, 220Geesec<strong>and</strong>idiasis, 317<strong>diseases</strong> caused by nontuberculousmycobacteria, 113relapsing fever, tick-borne, 272rhinosporidiosis, 351salmonellosis, 239tuberculous, 113Gibbons (see Primates, nonhuman)Gilchrist’s disease (see Blas<strong>to</strong>mycosis)Gl<strong>and</strong>ers, 142-145cutaneous, 142rodents (see Melioidosis)transmission, mode of, figure, 144Goats (see Caprines)Gorillas (see Primates, nonhuman)Guinea pigsanthrax, 26c<strong>and</strong>idiasis, 317corynebacteriosis, 101enterocolitis due <strong>to</strong> Clostridiumdifficile, 133, 135listeriosis, 173melioidosis, 186, 188plague, 208, 215rat-bite fever, 227yersiniosis, pseudotuberculous, 219,220, 223(see also Rodents)Gulls, 70, 240(see also Birds)HHamstersenterocolitis due <strong>to</strong> Clostridiumdifficile, 133, 135gl<strong>and</strong>ers, 144lep<strong>to</strong>spirosis, 163, 166salmonellosis, 241Hansen’s disease (see Leprosy)Hanseniasis (see Leprosy)Haplomycosis (see Adiaspiromycosis)Haplosporangiosis (seeAdiaspiromycosis)Haresbrucellosis, 42, 43, 47, 49-50derma<strong>to</strong>phy<strong>to</strong>sis, 335plague, 212, 214tuberculosis, zoonotic, 292tularemia, 275, 276, 277, 278, 279,280yersiniosisenterocolitic, 125pseudotuberculous, 219, 221, 222(see also Lagomorphs)Hippopotamuses, 23Hirudo medicinalis, 8His<strong>to</strong>plasma, 314, 339capsulatum, 339, 340, 342farciminosum, 354His<strong>to</strong>plasmosis, 339-345Horses (see Equines)Horseflies, tularemia, 276, 280IInfectionalgal (see Pro<strong>to</strong>thecosis)caused by Capnocy<strong>to</strong>phagacanimorsus, 146-148caused by Capnocy<strong>to</strong>phagacynodegmi, 146-148caused by DF-2 <strong>and</strong> DF-2–likebacteria (see Infection causedbyCapnocy<strong>to</strong>phaga canimorsus<strong>and</strong> C. cynodegmi)clostridial, of wounds, 87-89experimental, 45, 153his<strong>to</strong><strong>to</strong>xic (see Infection, clostridial,of wounds)listerial (see Listeriosis)natural, 41, 45, 47, 49, 50, 69, 150,151, 153, 181, 214, 276nontuberculous mycobacterial (seeDiseases caused by nontuberculousmycobacteria)nosocomial, 71, 130, 133strep<strong>to</strong>coccal (see Strep<strong>to</strong>coccosis)Infertility


INDEX 371brucellosis, 43, 45-47campylobacteriosis, 73, 76epizootic (see Diseases caused byCampyobacter fetus)lep<strong>to</strong>spirosis, 159, 161In<strong>to</strong>xicationVibrio parahaemolyticus, foodborne,138Ixodes, 179-181, 279dammini, 179-181holocyclus, 181pacificus, 179, 181persulcatus, 180, 181ricinus, 179, 181KKoalas, disease caused bynontuberculous mycobacteria, 112Lagomorphsplague, 212tularemia, 276, 277, 278, 279(see also Hares)Lama pacos, 49Lambs (see Ovines)Lamziekte (see Botulism)Leeches, aeromoniasis, 8, 9, 12Leprosy, 149-157Lep<strong>to</strong>sphaeria senegalensis, 345Lep<strong>to</strong>spiraballum, 161biflexa, 158, 164canicola, 158-162, 165grippotyphosa, 159-161, 165hardjo, 159, 160, 161, 163-165icterohaemorrhagiae, 158-162, 165interrogans, 158, 159, 163paidjan, 159, 161pomona, 158-161, 165-166pyrogenes, 161sejroe, 160tarassovi, 160, 161Lep<strong>to</strong>spirosis, 157-168transmission cycle, synanthropic,figure, 162Lepuscalifornicus, 279europaeus, 42, 47 ,49-50, 221, 279timidus, 276Lvariabilis, 279Leukocy<strong>to</strong>sis (see Listeriosis)Limberneck (see Botulism)Lion, 312Listerella monocy<strong>to</strong>genes (see Listeriamonocy<strong>to</strong>genes)Listeria, 168, 172, 175, 176ivanovii, 168, 173monocy<strong>to</strong>genes, 168-176Listeriasis (see Listeriosis)Listeriosis, 168-179Lizards (see Reptiles)Lockjaw (see Tetanus)Lucillia cuprina, 104Lyme disease, 179-184Lymphoreticulosis, benign, frominoculation (see Cat-scratchdisease)Lynx, 10MMacacaarc<strong>to</strong>ides, 112fascicularis, 185menestrina, 197mulatta, 197, 252nigra, 248silenus, 248sylvanus, 248Macaques (see Primates, nonhuman)Madura foot (see Myce<strong>to</strong>ma)Madurellagrisea, 345myce<strong>to</strong>matis, 345Maduromycosis (see Myce<strong>to</strong>ma)Maduromycotic myce<strong>to</strong>ma (seeMyce<strong>to</strong>ma)Mal rojo (see Erysipelas, animal <strong>and</strong>human erysipeloid)Maliasmus, 142Malleomycesmallei (see Pseudomonas mallei)pseudomallei (see Pseudomonaspseudomallei)Mammalsadiaspiromycosis, 303aeromoniasis, 7, 9aspergillosis, 305, 309blas<strong>to</strong>mycosis, 311botulism, 35, 36-37brucellosis, 49


372 INDEXcampylobacteriosis, 68, 69, 70c<strong>and</strong>idiasis, 316coccidioidomycosis, 320<strong>diseases</strong> caused by nontuberculousmycobacteria, 107, 109, 111erysipelas, animal, 14, 15his<strong>to</strong>plasmosis, 340lep<strong>to</strong>spirosis, 158listeriosis, 171, 173Lyme disease, 181-182nocardiosis, 195, 196pasteurellosis, 199, 200, 204plague, 211, 214salmonellosis, 236tuberculosis, zoonotic, 266, 287-294tularemia, 276Vibrio cholerae, non-O1, 118yersiniosisenterocolitic, 124pseudotuberculous, 219, 221, 223zygomycosis, 357(see also individual species)Margaropus decoloratus, 272Marmots, 212, 216Marsupialsplague, 214salmonellosis, 240Mas<strong>to</strong>mys natalensis, 49Meles meles, 292, 294Melioidosis, 184-190transmission, mode of, figure, 187Meli<strong>to</strong>coccosis (see Brucellosis)Mephitis mephitis, 303, 304Miceaeromoniasis, 7anthrax, 26botulism, 34c<strong>and</strong>idiasis, 317corynebacteriosis, 100derma<strong>to</strong>phy<strong>to</strong>sis, 335enterocolitis due <strong>to</strong> Clostridiumdifficile, 135leprosy, 151lep<strong>to</strong>spirosis, 162listeriosis, 173Lyme disease, 180plague, 215rat-bite fever, 226, 227relapsing fever, tick-borne, 273tularemia, 288yersiniosis, pseudotuberculous, 219,221(see also Rats <strong>and</strong> Rodents)Microsporum, 332, 333, 334, 337audouinii, 332, 334canis, 332-336equinum, 332, 335gallinae, 332gypseum, 333, 336nanum, 332, 336persicolor, 332vanbreuseghemii, 333Minksbotulism, 33, 36brucellosis, 50tuberculosis, zoonotic, 292tularemia, 276Mollusksaeromoniasis, 8, 11erysipelas, animal, 18poisoning caused by Vibrioparahaemolyticus, 139, 140, 141Moniliasis (see C<strong>and</strong>idiasis)Monkeyscampylobacteriosis, 69leprosy, 151, 152melioidosis, 185nocardiosis, 195, 197salmonellosis, 241shigellosis, 248, 249tuberculosis, zoonotic, 285, 291, 293,294yersiniosisenterocolitic, 126pseudotuberculous, 221(see also Primates, nonhuman)Monodelphis domestica, 214Mosqui<strong>to</strong>es, 276, 279Mucor, 357Mucorales, 307, 356Mucormycosis (see Zygomycosis)Mugil cephalus, 9Mustela nivalis, 304Myce<strong>to</strong>ma, 345-347eumycotic (see Myce<strong>to</strong>ma)maduromycotic (see Myce<strong>to</strong>ma)Mycobacteriosis (see Diseases causedby nontuberculous mycobacteria)Mycobacterium, 99, 107, 110, 153africanum, 107, 114, 283, 285, 288,291avium, 107, 108, 112, 113, 114, 115,288, 290


INDEX 373bovis, 107, 109, 111, 112, 114, 283-296chelonae, 108, 109fortuitum, 108-113intracellulare, 107, 108, 113kansasii, 108, 111leprae, 112, 149-155lepraemurium, 112marinum, 108, 110, 112, 113microti, 107, 283paratuberculosis, 107, 110, 111porcinum, 112scrofulaceum, 107, 109, 110simiae, 108, 109szulgai, 108, 109tuberculosis, 107, 108-111, 112, 113,114, 283, 285-291, 293-295ulcerans, 110, 112xenopi, 108, 111, 112, 113Mycoses, 303-360Myonecrosis (see Clostridial woundinfections)NNecrobacillosis, 190-195Neo<strong>to</strong>ma lepida, 41, 49Nocardia, 99, 195-198asteroides, 195-198, 345brasiliensis, 195-197, 345otitidiscaviarum, 195-197, 345Nocardiosis, 195-198Nontyphoid salmonellosis (seeSalmonellosis)North American blas<strong>to</strong>mycosis (seeBlas<strong>to</strong>mycosis)Nosocomialcase, 242infection, 130, 133, 136transmission, 127, 136, 238OOdocoileus virginianus, 180, 182Ohara’s disease (see Tularemia)Ondatra zibethicus, 276, 279Opossumsbrucellosis, 50Lyme disease, 180relapsing fever, tick-borne, 272tuberculosis, zoonotic, 289, 291, 294Ornithodoros, 271-274brasiliensis, 271erraticus, 271hermsii, 271, 273moubata, 271, 273rudis, 271, 273talaje, 273turicata, 271, 273venezuelensis (see O. rudis)verrucosus, 271O<strong>to</strong>mycosis, 307Ovinesanthrax, 24botulism, 33, 35brucellosis, 41, 42, 43, 48, 50, 53, 59,60, 61campylobacteriosis, 69, 70, 73, 74,75, 76c<strong>and</strong>idiasis, 317coccidioidomycosis, 321, 322colibacillosis, 95, 97corynebacteriosis, 100, 102cryp<strong>to</strong>coccosis, 328derma<strong>to</strong>philosis, 104, 105derma<strong>to</strong>phy<strong>to</strong>sis, 335<strong>diseases</strong> caused by nontuberculousmycobacteria, 107, 110erysipelas, animal, 15, 16-17food poisoning, clostridial, 83, 84, 85his<strong>to</strong>plasmosis, 340, 342infectioncaused by Capnocy<strong>to</strong>phagacanimorsus, 146, 147caused by Capnocy<strong>to</strong>phagacynodegmi, 146, 147of wounds, clostridial, 88lep<strong>to</strong>spirosis, 161listeriosis, 171, 172-173, 174, 175melioidosis, 185, 186, 188necrobacillosis, 191, 192, 193nocardiosis, 195pasteurellosis, 202, 203plague, 211, 213pro<strong>to</strong>thecosis, 349relapsing fever, tick-borne, 273rhodococcosis, 231salmonellosis, 238, 240strep<strong>to</strong>coccosis, 260tetanus, 266, 268tuberculosis, zoonotic, 289, 293, 294tularemia, 276, 278, 279, 280Vibrio cholerae, non-O1, 119yersiniosisenterocolitic, 126


374 INDEXpseudotuberculous, 220zygomycosis, 357, 358Oysters (see Mollusks)PPanthera leo, 312Paracoccidioides brasiliensis, 314Parakeets (see Birds)Paratyphi A, B, <strong>and</strong> C, 234Parrots (see Birds)aeromoniasis, 11tuberculosis, avian, 113Partridges (see Birds)Pasteurella, 199-202, 204caballi, 199canis, 199dagmatis, 199haemolytica, 199, 201, 202, 204mul<strong>to</strong>cida, 199-202, 203, 204pestis (see Yersinia pestis)septica (see P. mul<strong>to</strong>cida)s<strong>to</strong>matis, 199tularensis (see Francisella tularensis)x (see Yersinia enterocolitic)Pasteurellosis, 199-206Pediculus humanis, 214Pelicans (see Birds)Peromyscus leucopus, 180, 181, 212Pest (see Plague)Phascolarc<strong>to</strong>s cinereus, 112Pheasantsbotulism, 33, 37erysipelas, animal, 17Pigeons (see Birds)Pigs (see Swine)Plague, 207-218cases <strong>and</strong> deaths, figure, 209cases <strong>and</strong> deaths, Americas, table,210transmission cycle, domestic <strong>and</strong>peridomestic, figure, 213Pleurodemacinera, 112marmoratus, 112Pneumoniafibrinous (see Pasteurellosis)pasteurella (see Pasteurellosis)Pneumonomycosis (see Aspergillosis)Poikilotherms<strong>diseases</strong> caused by nontuberculousmycobacteria, 109, 112listeriosis, 171Poisoningbotulinum (see Botulism)foodcaused by Vibrioparahaemolyticus, 138-142clostridial, 82-87staphylococcal, 251-257Polygenis bohlsi jordani, 214Posada’s disease (seeCoccidioidomycosis)Poultsc<strong>and</strong>idiasis, 316, 317(see also Turkeys)Primates, nonhumancryp<strong>to</strong>coccosis, 328disease caused by nontuberculousmycobacteria, 110, 112leprosy, 151, 153, 154melioidosis, 185, 186, 187shigellosis, 248, 249, 250tuberculosis, 110zoonotic, 283, 285, 291, 293tularemia, 280Procyon lo<strong>to</strong>r, 180Proechimysguyanensis, 342semispinosus, 240Pro<strong>to</strong>theca, 348, 349wickerhamii, 348, 349zopfii, 348, 349Pro<strong>to</strong>thecosis, 348-350Pseudallescheria boydii, 345-346Pseudomonas, 9aeruginosa, 9mallei, 142pseudomallei, 184-188Pseudotuberculous yersiniosis, 218-225transmission, probable mode of,figure, 222Pulex irritans, 211, 214Pustule, malignant (see Anthrax)RRabbitsaeromoniasis, 7colibacillosis, 90, 94coryza (see Pasteurellosis)enterocolitis due <strong>to</strong> Clostridiumdifficile, 135fever (see Tularemia)


INDEX 375listeriosis, 171, 173melioidosis, 186pasteurellosis, 202-203snuffles (see Pasteurellosis)tularemia, 276, 277, 278, 279yersiniosis, pseudotuberculous, 221Raccoons, 180Rangifer caribou, 49Rat-bite fever, 226-229Ratsbrucellosis, 41, 49corynebacteriosis, 89derma<strong>to</strong>phy<strong>to</strong>sis, 334<strong>diseases</strong> caused by nontuberculousmycobacteria, 112lep<strong>to</strong>spirosis, 161, 162melioidosis, 186pasteurellosis, 203plague, 207, 208, 212, 214rat-bite fever, 226-229salmonellosis, 240tetanus, 268tularemia, 279yersiniosis, pseudotuberculous, 221(see also Mice <strong>and</strong> Rodents)Reindeer, brucellosis, 51Reptilesaeromoniasis, 8, 9, 10, 12listeriosis, 171rhodococcosis, 231salmonellosis, 236, 240, 241yersiniosis, pseudotuberculous, 219zygomycosis, 357, 359Reticuloendothelial cy<strong>to</strong>mycosis (seeHis<strong>to</strong>plasmosis)Rhamdia sapo, 9Rhinosporidiosis, 350-352Rhinosporidium seeberi, 350Rhipicephalusevertsi, 272sanguineus, 50Rhizomucor, 357, 358Rhizopus, 357, 358Rhodococcosis, 229-232Rhodococcus, 97equi, 176, 229-232, 289Ringworm (see Derma<strong>to</strong>phy<strong>to</strong>sis)Rodentia, 212, 275Rodentsbrucellosis, 49coccidioidomycosis, 321corynebacteriosis, 100, 101derma<strong>to</strong>phy<strong>to</strong>sis, 334-337<strong>diseases</strong> caused by nontuberculousmycobacteria, 107erysipelas, animal, 18lep<strong>to</strong>spirosis, 158, 161, 163, 165listeriosis, 171, 175Lyme disease, 181plague, 207, 211-216rat-bite fever, 226, 227relapsing fever, tick-borne, 272, 273,274salmonellosis, 240sporotrichosis, 353tuberculosis, zoonotic, 283tularemia, 275, 276, 277-280yersiniosisenterocolitic, 125pseudotuberculous, 221, 223(see also Mice <strong>and</strong> Rats)Ruminants (see individual species)SSaccharomyces neoformans (seeCryp<strong>to</strong>coccus neoformans)Saiga tatarica, 50Salmon, pro<strong>to</strong>thecosis, 349Salmonella, 55, 68, 83, 124, 233-244,250, 308abortus equi, 236, 237, 238, 243abortus ovis, 236, 238arizonae, 240, 241choleraesuis, 236-237, 240, 243dublin, 237, 238, 243enteritidis, 234-235, 237, 238, 240-244gallinarum, 233, 236, 239, 242, 243hadar, 244marina, 241oranienburg, 235poona, 240, 241pullorum, 233, 236, 237, 239, 242-243, 244sendai, 237thompson, 235typhi, 234, 236, 240, 242typhimurium, 233-235, 237-240,243-244weltevreden, 234Salmonellosis, 233-246outbreaks, selected countries, figure,235


376 INDEXtransmission, mode of, figure, 241San Joaquin Valley fever (seeCoccidioidomycosis)Scarlatina (see Strep<strong>to</strong>coccosis)Schmorl’s disease (see Necrobacillosis)Sciurus carolinensis, 180Sea lionsblas<strong>to</strong>mycosis, 312erysipelas, animal, 14Septicemia, hemorrhagic (seePasteurellosis)Sheep (see Ovines)Shiga-like, 90Shigella, 90, 124, 247-250boydii, 247, 248dysenteriae, 247, 248flexneri, 247, 248, 250sonnei, 247, 248, 250Shigellosis, 247-251Shrimp (see Mollusks)Skunksadiaspiromycosis, 303-304relapsing fever, tick-borne, 273Snakesaeromoniasis, 10salmonellosis, 240shigellosis, 249(see also Reptiles)Snuffles (see Pasteurellosis)Sodoku (see Rat-bite fever)Solipeds, gl<strong>and</strong>ers, 143-145Sparrows (see Birds)Sphaeroporus pseudonecrophorus, 190Spirillum (see Borrelia), 271minus, infection due <strong>to</strong>, 226, 227-228Spirochaeta (see Borrelia)Spiroche<strong>to</strong>sis (see Tick-borne relapsingfever)Spironema (see Borrelia)Sporothrix schenckii, 352-354Sporotrichosis, 352-356Sporotrichum beurmanni (seeSporothrix schenckii)Squirrelscorynebacteriosis, 101Lyme disease, 180plague, 212relapsing fever, tick-borne, 272yersioniosis, pseudotuberculous, 223(see also Rodents)Staphylococcal food poisoning, 251-257Staphylococcus, 251aureus,176, 251, 254, 259hyicus, 251intermedius, 251, 252Strep<strong>to</strong>bacillus moniliformis, infectiondue <strong>to</strong>, 226-227Strep<strong>to</strong>coccal sore throat (seeStrep<strong>to</strong>coccosis)Strep<strong>to</strong>coccosis, 257-265Strep<strong>to</strong>coccus, 257, 262acidominimus, 258agalactiae, 258-261, 263bovis, 258, 259canis, 258cremoris, 258dysgalactiae, 260, 263equi, 258, 259, 260, 263equisimilis, 259, 260lactis, 258mastitidis (see S. agalactiae)pyogenes, 258, 260, 261, 262suis, 257-260, 262-263uberis, 258, 260zooepidemicus, 258-261, 263Strep<strong>to</strong>myces somaliensis, 345, 346Strep<strong>to</strong>thrichosis (see Derma<strong>to</strong>philosis)Stuttgart disease (see Lep<strong>to</strong>spirosis)Swallows (see Birds)Swineactinomycosis, 4aeromoniasis, 11anthrax, 24, 26botulism, 33brucellosis, 42, 47, 48, 50, 53, 58, 61campylobacteriosis, 70c<strong>and</strong>idiasis, 317coccidioidomycosis, 321colibacillosis, 95, 96, 97derma<strong>to</strong>phy<strong>to</strong>sis, 336disease caused by nontuberculousmycobacteria, 107, 109, 111, 112erysipelas, animal, 14, 15, 16, 17, 18,19food poisoningclostridial, 84, 85staphylococcal, 253lep<strong>to</strong>spirosis, 158, 160, 162, 165, 166listeriosis, 171, 173melioidosis, 185, 186, 188necrobacillosis, 192pasteurellosis, 202rhodococcosis, 231salmonellosis, 238, 240-241


INDEX 377sporotrichosis, 353strep<strong>to</strong>coccosis, 257, 259, 260, 261,262tuberculosis, 111zoonotic, 285, 287, 288-289, 293,294tularemia, 278yersiniosisenterocolitic, 124, 125, 126-127,129, 130pseudotuberculous, 221-222zygomycosis, 58Sylvilagus, tularemia, 275, 279Syndrome, cat-scratch (see Cat-scratchdisease)Tamias striatus, 180Tetanus, 265-271morbidity in Argentina, table, 267Thrush (see C<strong>and</strong>idiasis)Tick-borne relapsing fever, 271-274transmission, mode of, figure, 273Ticksbrucellosis, 50derma<strong>to</strong>philosis, 104, 105Lyme disease, 179-182relapsing fever, tick-borne, 271-274tularemia, 275-276, 278-281Tinea (see Derma<strong>to</strong>phy<strong>to</strong>sis)Torula his<strong>to</strong>lytica (see Cryp<strong>to</strong>coccusneoformans)Torulopsis neoformans (seeCryp<strong>to</strong>coccus neoformans)Torulosis (see Cryp<strong>to</strong>coccosis)Toucan (see Birds)Toxicosisclostridial (see Clostridial foodpoisoning)staphylococcal alimentary (seeStaphylococcal food poisoning)Trichomonas, 316Trichophy<strong>to</strong>n, 332, 333, 334, 335, 337album, 335discoides, 335equinum, 335erinacei, 332faviforme, 335gallinae, 332, 336gypseum, 333, 336mentagrophytes, 332-336Tochraceum, 335rubrum, 333, 334, 336simii, 332verrucosum, 332-333, 335, 336, 337Tripanscorax fragilecus, 50Trismus (see Tetanus)Tuberculosis, zoonotic, 283-299transmission, mode of, figure, 293Tularemia, 275-282transmission, mode of, in theAmericas, figure, 279Tunga penetrans, 268Turkeysaeromoniasis, 11aspergillosis, 306, 308, 309campylobacteriosis, 70colibacillosis, 96<strong>diseases</strong> caused by nontuberculousmycobacteria, 113erysipelas, animal, 17food poisoning, staphylococcal, 253rat-bite fever, 226-227salmonellosis, 240tuberculosis, 113yersiniosis, pseudotuberculous, 219,220, 223Turtles, salmonellosis, 240-241Typhus, recurrent (see Tick-bornerelapsing fever)VVaccinesaeromoniasis, 12anthrax, 26-27botulism, 42brucellosis, 42, 53, 55-62c<strong>and</strong>idiasis, 319coccidioidomycosis, 324colibacillosis, 97<strong>diseases</strong> caused by Campylobacterfetus, 76erysipelas, animal, 16, 18, 19food poisoning, clostridial, 86, 89infection of wounds, clostridial, 89lep<strong>to</strong>spirosis, 165-166necrobacillosis, 193pasteurellosis, 199, 204rhodococcosis, 232salmonellosis, 243-244shigellosis, 250tetanus, 269-270


378 INDEXtuberculosis, zoonotic, 296tularemia, 281Vibriocholerae, 93cholerae non-O1, <strong>diseases</strong> in man<strong>and</strong> animals, 117-122fetus (see Campylobacter fetus)parahaemolyticus, 138-142Vibriosis (see Diseases caused byCampylobacter fetus; seeCampylobacteriosis)bovine genital (see Diseases causedby Campylobacter fetus; seeCampylobacteriosis)WWeasels, 272Weil’s disease (see Lep<strong>to</strong>spirosis)Whitmore’s disease (see Melioidosis)XXenopsylla cheopis, 188, 214YYaksbrucellosis, 49pasteurellosis, 201Yersiniaenterocolitica, 55, 122-132, 223pestis, 207, 211, 212, 214, 215pseudotuberculosis, 207, 218-223Yersiniosisenterocolitic, 122-132transmission, supposed mode of,figure, 127pseudotuberculous, 218-225transmission, probable mode of,figure, 222Zebras (see Equines)Zoonotic tuberculosis, 283-299transmission, mode of, figure, 293Zygomycetes, 356Zygomycosis, 356-360Z

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!