Open access peer-reviewed chapter

Diverse Bioactive Molecules from the Genus Lactobacillus

Written By

Rodney H. Perez and Amily E. Ancuelo

Submitted: 11 November 2021 Reviewed: 18 January 2022 Published: 12 June 2022

DOI: 10.5772/intechopen.102747

From the Edited Volume

Lactobacillus - A Multifunctional Genus

Edited by Marta Laranjo

Chapter metrics overview

241 Chapter Downloads

View Full Metrics

Abstract

Lactobacilli are widespread microorganisms and are broadly employed in a variety of applications. It is one of the LAB genera that has been designated as Generally Regarded as Safe (GRAS) and many of its member species are included in the Qualified Presumption of Safety (QPS) list. Lactobacillus is commonly utilized as a starter culture in many fermented food products, probiotics, and has long been used as natural bio-preservatives to increase shelf life and improve food quality and safety. Aside from the many benefits, it delivers in the food sector, the use of lactobacillus strains in the clinical setting as a prophylactic and/or treatment for a variety of diseases has gained increasing attention. These uses of lactobacillus are all made possible through the diverse bioactive molecules it generates. Lactobacillus exerts its positive health and nutritional effects through a variety of mechanisms, including inhibition of pathogen adhesion or colonization, metabolic activity through the synthesis of metabolites and enzymes, and immune system modulation among others. The ability of many lactobacillus strains to mediate the bio-conversion of certain metabolites has also been shown in numerous studies. This chapter describes the recent findings on the impact of the diverse bioactive molecules produced by different lactobacillus strains, their mode of action, and their application in different industries.

Keywords

  • lactic acid bacteria
  • GRAS
  • lactobacillus
  • bioactive compounds
  • probiotics

1. Introduction

One of the most significant, and extensively used lactic acid bacteria (LAB) is lactobacillus. This genus comprises a large number of species that can be found in diverse environments, such as in plants, food products, and mucosal surfaces of the human body. Lactobacilli are characterized by the formation of lactic acid as the primary end product of carbohydrate metabolism. Species of lactobacillus are Gram-positive, homofermentative, thermophilic, and non-spore-forming rods. Lactobacillus species ferment a broad array of carbohydrates and can ferment extracellular fructans, starch, or glycogen depending on the strain. All organisms formerly assigned to the Lactobacillus delbrueckii group are now included in the emended description of the genus [1].

Among LAB, lactobacillus is one of the genera considered as Generally Regarded as Safe (GRAS) by the U.S. Food and Drug Administration (FDA) as they are a safe means to generate products for a variety of industries. The European Food Safety Authority (EFSA) also grants Qualified Presumption of Safety (QPS) status to many lactobacillus species. The EFSA notes that some LAB strains are susceptible to acquiring virulence and antibiotic resistance genes and have opportunistic properties and hence excluded in the QPS list [2]. Lactobacilli are broadly applied in the food industry as both technical starters in fermented goods and probiotics due to their unique health and nutritional benefits [3, 4]. These microorganisms play an indispensable role in many foods fermentation. For instance, many lactobacilli species have been identified as primary microorganisms in sauerkraut fermentation [5]. Lactobacilli also play a significant part in malolactic fermentation, critical in winemaking [6]. In Chinese Maotai-flavored liquor production, lactobacillus accelerates flavor component conversion from alcohol (ethanol) to acid (lactic acid and acetic acid) [7]. Lactic acid fermentation using L. plantarum was found optimal in improving pea protein isolates aroma and taste [8]. Fermentation with lactobacillus also improved the foaming capabilities of egg white [9]. Many lactobacillus strains are also important in the manufacture of many fermented meat and meat products. Moreover, probiotic lactobacillus was given special attention as well for its ability to stimulate or modulate the immune system [10]. It also has possible applications in health-related areas such as intestinal inflammation [11], prevention of urinary tract infection [12], and treatment against cancer cells [13].

Indigenous LAB is constantly exposed to extreme conditions such as varying temperature, pH, and nutrient levels [14]. As a result, native LAB has been linked to higher competitive metabolic capacities, which encourage their growth as a competitive microbiota for other microorganisms in their natural habitat. The synthesis of a large number of bioactive metabolites is one of these capacities. Among LAB genera, lactobacilli are known producers of diverse bioactive molecules that offer a wide range of benefits to food, agricultural, industrial and clinical fields. They have long been exploited in food and animal feed as natural preservatives. Their antimicrobial action is mostly due to the production of organic acids, hydrogen peroxide, inhibitory compounds, as well as competition for nutrients and the development of antimicrobial compounds like bacteriocin [15]. Several studies have shown that the organic acids produced by lactobacillus like lactic acid kill pathogens at sufficient concentrations, such as Campylobacter jejuni by disrupting its membrane [16]. It also secretes ethanol and fatty acid as antimicrobial molecules. It can produce acetic acid, formic acid, and other acids [17].

Lactobacilli exhibit their beneficial properties through a wide range of processes that include a large spectrum of bioactive compounds. In this chapter, the utility of these microorganisms and their bioactive compound by-products for the promotion of better health and nutrition are summed up. Lactobacilli and their by-products can be utilized in technology and product development geared towards sustainable approaches for the improvement of human conditions targeted by the United Nations 2030 Agenda and its Sustainable Development Goals. The well-established functions of lactobacilli and their bioactive molecules in food fermentation could play a key role in ensuring that people around the world have access to safe and nutritious food by improving the current food production, safety, and preservation. Its application in the clinical setting also has the potential to address major health concerns, including sexual, reproductive, newborn, and environmental diseases which will be discussed in the following sections. Thus, this chapter will center the attention on the different bioactive molecules from the genus lactobacillus, such as bacteriocins, bioactive peptides, SCFAs, vitamins, enzymes, EPs, immune-modulating compounds, bio-converted molecules, and its probiotic properties.

Advertisement

2. Bioactive compounds produced by lactobacillus

2.1 Bacteriocins

Bacteriocins are multifunctional, ribosomal-synthesized antimicrobial peptides. The bactericidal activity of bacteriocins is demonstrated against species that are closely related to the producer strain [18]. The bactericidal or bacteriostatic actions of bacteriocins produced by Gram-positive bacteria, including LAB, are mostly against Gram-positive bacteria including food-borne pathogens [19]. Bacteriocins inhibit their target cells by destabilizing the bacterial cell membrane and/or creating pores resulting in the death of the target cells through a fast-acting mode of action that is active even at very low concentrations [18]. Bacteriocins from Gram-positive bacteria are divided into three classes based on their structural and physicochemical properties: class I (lantibiotics), which are lanthionine-containing peptides; class II, comprise the non-lanthionine-containing bacteriocins [20].

The promise of bacteriocins, particularly from lactic acid bacteria, for various applications has instigated a great deal of interest in bacteriocin research. LAB bacteriocins are recognized for their activity over a wide pH range and are inherently tolerant to extreme thermal stress. The fact that these antimicrobial peptides are colorless, odorless, and tasteless, adds to their potential uses [18]. Bacteriocins also offer a number of advantages over traditional antibiotics. The most notable of which is that they are primary metabolites with relatively straightforward biosynthetic processes compared to conventional antibiotics, which are secondary metabolites. Thus, bioengineering may readily improve their activity or specificity towards their target bacteria [18].

Lactobacillus, is a LAB genus that has been shown to produce diverse bacteriocins (Table 1). A lactobacillus strain isolated from traditional Egyptian dairy products showed antimicrobial actions against different Gram-positive and Gram-negative bacteria [35]. The highest inhibitory activity of L. brevis (B23) was exhibited against Escherichia coli, Staphylococcus and Bacillus.

ClassBacteriocinLactobacillus strainReference
IParaplantaricin TC318L. paraplantarum OSY-TC318[21]
Plantaricin CL. plantarum LL441[22]
Lactocin SL. sakei L45[23]
IIaPlantaricin 423L. plantarum 423[24]
Plantaricin LPL-1L. plantarum LPL-1[25]
Rhamnocin 519L. rhamnosus CJNU 0519[26]
IIbGassericin SL. gasseri LA327[27]
Gassericin T
Gassericin ML. gasseri LM19[28]
IIcAcidocin BL. acidophilus M46[29]
Plantaricyclin AL. plantarum NI326[30]
Plantacyclin B21AGL. plantarum WCFS1[31]
IIdSakacin D98aL. sakei D98[32]
Sakacin D98c
Bactofencin AL. salivarus DPC6502[33]
IIIHelveticin-ML. crispatus[34]

Table 1.

Diverse bacteriocins produced by various strains of Lactobacillus.

A lactobacillus strain has also been described to produce multiple bacteriocins. Lactobacillus sakei 5 from malted barley was found to produce three bacteriocins [36]. Genetic and functional analysis revealed that this strain generates a plasmid-encoded bacteriocin sakacin P, as well as two novels, chromosomally encoded bacteriocins, sakacin T and sakacin X. This strain may be a viable candidate for usage in the brewing sector since it inhibits bacterial strains known to cause severe spoiling problems in this industry.

A strain isolated from human breast milk, Lactobacillus gasseri LM19, was also found to produce several bacteriocins, including a novel bacteriocin, gassericin M [28]. In a complex environment that mimicked human colon circumstances, L. gasseri LM19 not only survived but also expressed seven bacteriocin genes and generated short-chain fatty acids. The gut origin of L. gasseri LM19 enabled it to thrive in GI tract conditions and display antagonistic properties against other gut bacteria, such as enteropathogens [28]. Different bacteriocin-producing L. plantarum strains have also been found in a variety of foods, including meat [37], fermented milk [38], cheese [39], and sourdough [40].

Aside from its usefulness in the food industry, bacteriocin-producing lactobacillus was also investigated for its use in the clinical setting, particularly in preventing and treating vaginal disorders. Lactobacillus is bacteria naturally found in the healthy human vagina [41] and urethra [42]. A low count of lactobacillus is inversely related to high numbers of E. coli in the vagina and a history of recurrent urinary tract infection [43]. A new bacteriocin generated by L. acidophilus KS400 was identified and characterized, as well as its antimicrobial properties against urogenital pathogens [44]. These species have been shown to colonize the epithelial surface and release antimicrobial compounds that regulate the vaginal microflora.

2.2 Bioactive peptides

Digestive proteases and peptidases from human’s release food-encrypted bioactive peptides that can be absorbed by the gut and then reach peripheral organs. However, the enzymatic activity of LAB largely contributes to their release, either into the food matrix or in the gut. Due to the limited length of the overall genome, the biosynthetic abilities of LAB are very limited especially in amino acid synthesis [45]. Therefore, LAB evolved a complex and sophisticated proteolytic system allowing them to get amino acids from the proteins present in the external environment [46]. The proteolytic system of LAB converts protein substrates into free amino acids and small peptides, which enables them to carry out their intrinsic physiological mechanisms such as regulation of intracellular pH, production of metabolic energy, stress tolerance, and biosynthesis of proteins [47].

Numerous bioactive peptides lack activity when protein is encrypted, but display their interesting biological functions when released proteolytically. They have been shown to hold health-promoting qualities as antimicrobials, hypocholesterolemic, opioid antagonists, angiotensin-converting enzyme inhibitors, anti-thrombotic, immuno-modulators, cytomodulators, and antioxidants [48]. The utilization of LAB such as lactobacillus in the synthesis and valorization of new bioactive peptides is a useful method. The proteolytic activity of lactobacillus is strain- and species-dependent: each species has a distinct proteinase composition, encompassing a wide range of proteolytic activities [49].

Over the past years, lactobacillus species have presented great potential as producers of bioactive peptides through fermentation using different protein matrices (Table 2). LAB proteolytic system is capable of producing bioactive peptides from a variety of food proteins, particularly casein, which is the major nitrogen source in their environment. [59]. L. helveticus CICC6024 was employed to effectively ferment milk-casein under fixed fermentation processes to facilitate an efficient bioactive peptide synthesis [60].

Lactobacillus strainBioactive peptidesProtein SourceReference
L. caseiAntihypertensive peptidesMilk[50]
L. caseiAntimicrobial, antioxidant and ACE-inhibitory peptidesWhey and skim milk[51]
L. plantarum
L. rhamnosus
L. helveticusAntioxidative, opioid, stimulating, hypotensive, immunomodulating, antibacterial, and antithrombotic peptidesκ-casein[52]
L. helveticusACE-inhibitory peptidesMilk[53]
L. helveticus R0389ACE-inhibitory and immunomodulating peptidesCasein[54]
L. rhamnosus R0011
Lactobacillus GGImmunostimulatory, opioid, and ACE-inhibitory peptidesUHT Milk[55]
L. plantarum 55Anti-inflammatory, antihemolytic, antioxidant, antimutagenic, and antimicrobial peptidesMilk[56]
L. plantarum C2Anti-oxidant and ACE-inhibitory peptidesSoy milk[57]
L. sanfranciscensis I4Anti-oxidant and anti-inflammatory peptidesItalian sourdough[58]
L. farciminis A19
L. rossiae A20

Table 2.

Bioactive peptides generated by Lactobacillus using different protein matrices.

Other lactobacillus strains have also been found to be capable of releasing bioactive peptides from food proteins. Milk inoculated with L. helveticus and casein hydrolysates generated by L. helveticus CP790 extracellular proteinase both contain antihypertensive peptides [61]. Antihypertensive substances were also recovered from an L. casei cell extract used in fermented milk production [50]. Two fermented kinds of milk containing ACE-inhibitory peptides were generated using L. delbrueckii subsp. bulgaricus and Lactococcus lactis subsp. cremoris strains [62]. Bioactive peptides have been discovered in UHT milk fermented by the probiotic Lactobacillus GG strain and digested by pepsin and trypsin enzymes. These bioactive peptides exhibit varying degrees of immunostimulatory, opioid, and ACE-inhibitory properties [55]. Cultures of L. plantarum, L. casei, and L. rhamnosus from a fecal sample of a human infant were employed as a proteolytic starter culture for the fermentation of skim milk and whey to release small peptides that have antimicrobial, antioxidant, and ACE inhibitory activities. These encrypted bioactive peptides can be utilized as a functional food and/or, dietary supplement, to provide particular health advantages.

Single-activity milk-derived bioactive peptides have been widely reported. The anti-inflammatory, antihemolytic, antioxidant, antimutagenic, and antimicrobial activities of crude extracts and peptide fractions obtained from fermented milk with specific L. plantarum strains were assessed [56]. L. plantarum 55 was found to generate encrypted peptides with extensive capabilities as dietary bioactive components for the development of nutraceutical biotechnological products.

2.3 Short-chain fatty acids (SCFAs)

The small and large intestines of humans lack several carbohydrate-digesting enzymes that can be produced by probiotic bacteria. However, the probiotic bacteria ferment these undigested carbohydrates and produce energy that is utilized by the host to carry out various functions. The undigested sugars are converted into short-chain fatty acids (SCFAs) such as butyrate, acetate, and propionate. The typical reaction of SCFAs production and overall stoichiometry has been summarized and is shown as follows [63]:

59C6H12O6+38H2O60CH3COOH+18CH3CH2CH2COOH+22CH3CH2COOH+96CO2+134H2+HeatE1

SCFAs from lactobacillus have been proven to have therapeutic effects against several diseases through their antimicrobial potential (Table 3). For example, L. reuteri produces SCFAs to inhibit colon cancer cell proliferation [69]. Several L. reuteri strains were shown to synthesize SCFAs and demonstrated growth inhibitory activity against colorectal cancer cells. Thus, the anti-cancer action and the ability to generate anti-carcinogenic active substances of L. reuteri indicate that it may be used as a bio-therapeutic. Moreover, the impact of L. paracasei CNCM I-1572 on clinical and gut microbiota-related parameters in irritable bowel syndrome (IBS) was also investigated [65]. L. paracasei CNCM I-1572 was shown to regulate the structure and function of gut microbiota and decrease immunological activation in IBS by substantially increasing the SCFAs acetate and butyrate and a corresponding decrease in the pro-inflammatory cytokine interleukin-15. Several lactobacillus strains were also investigated for their application to treat bacterial vaginosis [64]. The strain L. plantarum ZX27 was found to produce more short-chain fatty acids and lactic acid and inhibited Gardnerella vaginalis growth and adherence.

Lactobacillus strainSCFASpectrumReference
L. delbrueckii DM8909Acetatic acid, propionic acid, butyric acid, isobutyric acid, valeric acid, and isovaleric acidGardnerella vaginalis[64]
L. plantarum ATCC14917
L. plantarum ZX27
L. paracasei CNCM I-1572Acetatic acid and butyric acidRuminococcus bromii and Ruminococcus callidus[65]
L. paracasei subsp. paracasei NTU 101Acetic acid, propionic acid, and butyric acidClostridium perfringens and Enterobacteriaceae[66]
L. plantarum G72Formic acid, acetic acid, propionic acid, and butyric acidListeria monocytogenes, Salmonella typhimurium, E. coli, and S. aureus[67]
L. rhamnosusAcetic acid, propionic acidE. coli[68]
L. reuteri NCIMB -11,951, −701,359, −701,089, −702,655, and − 702,656Acetic acid, propionic acid, and butyric acidCaco-2 colon cancer cells[69]
L. salivarius ssp. salicinius JCM 1230Propionic acid, and butyric acidSalmonella[70]
L. agilis JCM 1048

Table 3.

Antimicrobial spectrum of the SCFAs produced by Lactobacillus strain.

SCFAs are generated by bacteria in the gastrointestinal system, which relies on non-digestible carbohydrates for energy. SCFA production is necessary to increase the acidity of the gut environment, which inhibits many harmful microorganisms. Production of SCFAs has been shown as one mechanism of lactobacillus strains to inhibit the development of metabolic syndrome by its influence on microbiota modulation [71]. The production of SCFAs and antimicrobial activity of L. plantarum G72 for its potential application in improving the diet of pregnant women.

2.4 Vitamins

Vitamins are essential micronutrients that are required for the metabolism of every organism. Humans are incapable of producing vitamins, resulting in vitamin deficiencies, malnutrition, and stunted growth from infants to the elderly. Thus, they must be acquired exogenously (i.e., in the form of diet). All vitamins can be classified into two groups: water-soluble vitamins and fat-soluble vitamins. Water-soluble B-group vitamins are generated by several bacteria and are consumed in the gut. Fat-soluble vitamins, on the other hand, are taken in the digestive tract using lipids as micelles. Plants and animals are natural providers of vitamins, although certain vitamins are chemically produced.

Lactic acid bacteria, especially lactobacillus, are known to be good producers of vitamins. Lactobacillus decreases the overall growth of bacteria-caused diseases by generating these nutritional components (Table 4). Lactobacillus strains from traditional yogurt were able to produce B-group vitamins [73]. L. paracasei subsp. tolerance JCM 1171 (T), L. acidophilus KU, and L. fermentum showed the highest amount of Vitamin B6 and B9, B3, and B2, respectively. L. plantarum LZ95 originally from infant feces and CY2 from fresh milk were identified to be capable of producing a high level of extracellular vitamin B12 as well [75]. Moreover, co-fermentation of glycerol and fructose in soy-yogurt by L. reuteri has been demonstrated to enhance vitamin B12 synthesis [77].

Lactobacillus strainVitaminsReference
L. fermentum KGL2Vitamin B2, B9, and B12[72]
L. plantarum KGL3A
L. fermentum KGL4
L. rhamnosus RNS4
L. fermentum WTS4
L. paracasei subsp. tolerance JCM 1171TVitamin B2, B3, B6, and B9[73]
L. acidophilus KU
L. fermentum
L. plantarum CRL2130Vitamin B2[74]
L. plantarum LZ227Vitamin B2 and B9
L. plantarum LZ95Vitamin B12 (adenosylcobalamin and methylcobalamin)[75]
L. plantarum CY2
L. plantarum BHM10Vitamin B12[76]
L. plantarum BCF20
L. reuteriVitamin B12[77]
L. rossiae DSM 15814TVitamin B12[78]

Table 4.

Example of Lactobacillus strains known to produce vitamins.

Some vitamins, particularly riboflavin and folate derivatives, have been shown to help combat certain diseases. Vitamin-producing lactic acid bacteria, particularly strains that produce folate and riboflavin in combination with immune-stimulating strains, could be used as effective alternative types of treatment in patients suffering from a variety of inflammatory diseases [79]. Riboflavin-producing L. plantarum CRL2130, through oral administration, exhibited its ability to prevent trinitrobenzene sulfonic acid-induced colitis in mice, reducing pro-inflammatory cytokines [74].

2.5 Enzymes

Lactic acid bacteria perform metabolic processes due to the synthesis of enzymes. Enzymes play a critical role in biological reactions by acting as biocatalysts, mediating all anabolic and catabolic pathways, and lowering the activation energy of biochemical reactions. The digestive enzymes in the lysosomes, for example, enhance the digestion of a wide range of substances absorbed from outside the cell in the gastrointestinal tract (GIT). These enzymes work together to convert carbohydrates, proteins, and lipids into monomers that can be absorbed by human cells. Examples of digestive enzymes include amylase, lactase, pepsin, trypsin, pancreatic amylase, lipase, nuclease, maltase, and lactase [80].

Lactobacillus strains are well-known enzyme producers (Table 5). Amylases are one of the most often utilized enzymes in industry. These enzymes hydrolyze starch molecules into polymers made up of glucose units [90]. Lactobacillus amylases are considered safe since they are non-pathogenic and the end product of their fermentation is lactate, a commonly utilized flavoring ingredient in the food industry [91]. Several lactobacillus strains such as L. brevis, L. casei, and L. fermentum, were shown to produce a significant quantity of amylase [81]. The amylolytic potential of lactobacillus strains from wet-milled cereals, cassava flour, and fruits has been studied. L. plantarum (AMZ5) showed amylolytic potential through starch hydrolysis as it exhibited remarkable starch degradation capacity. [92].

Lactobacillus strainEnzymesReference
L. brevisAmylase[81]
L. fermentum
L. brevis C10CpSA3b6β-glucanase[82]
L. crispatus I12pSA3b6
L. brevis I23pSA3b6
L. fermentum I25pSA3b6
L. brevis I211pSA3b6
L. brevis I218pSA3b6
L. bulgaricusβ-galactosidase enzyme[83]
L. caseiAmylase and invertase[81]
L. casei LFTI® L26ACE-inhibitory enzyme[84]
Lactobacillus delbrueckii QS306ACE-inhibitory enzyme[85]
L. helveticus IMAU80872, IMAU80852, and IMAU80851ACE-inhibitory enzyme[86]
L. plantarumα-galactosidase enzyme[87]
Lactobacillus rhamnosusβ-galactosidase enzyme[88]
Lactobacillus sp. G3_4_1TO2Amylase[89]

Table 5.

Example of enzyme-producing Lactobacillus strains.

Angiotensin-converting enzyme (ACE, EC 33.4.15.1, CD143) has a significant impact on the regulation of arterial blood pressure [93]. Inhibiting this enzyme can cause antihypertensive effects. Because of its role in the renin-angiotensin and kinin-nitric oxide systems, ACE-inhibitors are an ideal physiological target for clinical hypertension treatment [86]. However, ACE inhibitors that are currently available are synthetic pharmacological medicines that are not recommended for usage in healthy or low-risk populations due to side effects such as dry cough, skin rashes, and angioneurotic edema. As a result, producing safe and natural ACE inhibitors is critical for future hypertension therapy and prevention [94]. Previous studies show that ACE inhibitors are already been isolated from different products such as milk [95], cheese [96], yogurt [84], and other dairy products. The L. helveticus strains IMAU80872, IMAU80852, and IMAU80851 from fermented milk possessed a high ACE-inhibitory activity [86]. ACE inhibitory peptides were also isolated and identified from milk fermented with L. delbrueckii QS306 [85]. Moreover, ACE-inhibitory peptides account for the majority of bioactive peptides generated during yogurt fermentation processes. A strong link between L. casei LFTI® L26 growth and ACE inhibition in all yogurt samples was discovered during the initial stages of storage, compared to control yogurt, which reduced substantially after storage [84]. These previous researches prove that bioactive ACE-inhibitory producing lactobacillus strains have a great deal of potential for the improvement and production of functional dairy food products with antihypertensive effects.

The β-galactosidase enzyme, one of the glycosidases, is widely used in the dairy industry as well. These are produced by most lactobacillus species. Lactose, the primary carbohydrate in milk, is hydrolyzed by this enzyme into glucose and galactose, which may be absorbed via the intestinal epithelium. β-galactosidase involves two enzymatic activities: one hydrolyzes lactose and also cleaves cellobiose, cellotriose, cellotetrose, and to some extent cellulose, while the other splits β-glycosides [97]. High β-galactosidase activity observed in L. rhamnosus [88] and L. bulgaricus [83] has also been reported.

2.6 Exopolysaccharides (EPs)

Exopolysaccharides (EPs) are high–molecular, long-chain linear biopolymers containing side chains of homopolysaccharide or heteropolysaccharide carbohydrate units linked with α-glycosidic and β-glycosidic bonds [98]. The enzymes such as glycosyltransferase and glycoside hydrolase convert the sugar nucleotide precursors into EPs. EPs are “food-grade biopolymers,” or extracellular biopolymers with a high molecular weight that are acquired from natural sources and produced during the metabolism of microorganisms [99].

Lactobacillus is one of the species of LAB that is frequently regarded as EPs-producing microorganisms (Table 6). An EPs termed as LPC-1 from L. plantarum C88 showed strong antioxidant activity and exhibited strong hydroxyl radical scavenging activity [107]. A novel EPs was also isolated from L. plantarum KX041 culture from a traditional Chinese pickle juice sample [109]. The EPs had a molecular weight of 38.67 KDa, which exhibited high thermal stability. EPS generated by L. plantarum has a good prospect to be utilized as natural antioxidants or functional additives in the food sector.

Lactobacillus strainExopolysaccharideReference
L. delbrueckii ssp. bulgaricus SRFM-1Glucose and galactose[100]
L. gasseri FR4Glucose, mannose, galactose, rhamnose and fucose[101]
L. helveticus MB2–1Glucose, mannose, galactose, rhamnose, and arabinose (c-EPS)[102]
L. kefiri MSR101Galactose and glucose (MSR101)[103]
L. mucosae (DPC) 6426β-glucan[104]
L. pentosus 14FEGlucose[105]
L. pentosus 68FE
L. plantarum 47FE
L. plantarum 301102SGlucose and mannose[106]
L. plantarum C88Galactose and glucose (LPC-1)[107]
L. plantarum H31Glucose and mannose[108]
L. plantarum KX041Arabinose, mannose, glucose, and galactose[109]

Table 6.

Example of exopolysaccharides produced by Lactobacillus strains.

There has been a growing interest in using EPs-producing LAB for a variety of biological purposes. Among them, the anticancer action of EPs has attracted increasing attention. The EPs produced by L. kefiri MSR101 (MSR101 EPS) and its ability to inhibit the growth of HT-29 colon cancer cells were explored [103]. Structural analysis showed that MRS101 EPS is a heteropolysaccharide having a repeating unit of glucose and galactose and has a partial crystalline nature. In-vitro anticancer tests also showed significant anticancer action of MRS101 EPS against HT-29 cells. Moreover, a novel cell-bound exopolysaccharide (c-EPS) isolated from L. helveticus MB2–1 [102] showed high structural stability and may be used to make films and edible nanostructures for drug and food additive encapsulation. In vitro anticancer testing revealed that c-EPS exhibited substantial anticancer effects against human HepG-2 liver cancer, BGC-823 gastric cancer, and notably HT-29 colon cancer cells.

The utilization of probiotic microorganisms has been linked to a lower risk of cardiovascular disease, the leading cause of mortality and disability. The effect of dietary treatment of exopolysaccharide-producing probiotic lactobacillus on lipid metabolism and gut microbiota was investigated using apolipoprotein E (apoE)–deficient mice [104]. Dietary supplementation with a β-glucan–producing probiotic strain L. mucosae Dairy Product Culture Collection (DPC) 6426 resulted in lipid metabolism regulation in the mouse model of atherosclerosis. Several strains of L. delbrueckii subsp. bulgaricus isolated from homemade yogurt were also shown to produce EPs that can help cholesterol reduction [110]. The cholesterol removal mechanism, which involves binding or adhering to the bacterium cells, particularly to the EPS generated by the bacteria and enclosing the bacterial cells as a capsule, may be useful and relevant in human serum cholesterol management.

2.7 Immune-modulating compounds

Different lactobacillus strains synthesize immune-modulating compounds that confer various health effects (Table 7). These most widely utilized probiotic agents promote intestinal microbiota and gut health and regulate the immune system in consumers. The immune system is modulated by probiotic bacteria, which control the synthesis of antibodies, interleukins, cytokines, and lymphocytes [121]. The probiotic bacteria interact with intestinal epithelial cells and generate immunomodulatory molecules, which activate the host immune response. By stimulating the production of interleukin-10 (IL-10) and immunoglobulin A antibodies (IgA), probiotics regulate immunity and inflammatory gene expression, reducing the host immunological response to infections [122]. IgA production, which is stimulated by dendritic cells, naive T cells, and B cells, promotes immune-modulatory effects as well and helps to eliminate pathogenic bacteria.

Lactobacillus strainImmune-modulating compound/mechanismReference
L. acidophilus DSM 32241Increase in IgA and IgG levels[111]
L. helveticus DSM 32242
L. paracasei DSM 32243
L. plantarum DSM 32244
L. brevis DSM 27961
L. brevis B13–2Activated RAW 264.7 murine macrophages[112]
L. fermentum BioE LF11Inhibited secretion of lipopolysaccharide-induced pro-inflammatory cytokines IL-6 and TNF-α in RAW264.7 macrophages in vitro[113]
L. plantarum BioE LPL59
L. paracasei BioE LP08
L. fermentum CECT5716Modulation of intestinal cytokines IL-10 and IL-12[114]
L. fermentum JDFM216Increased sIgA[115]
L. gasseriIncreased IFN-levels[116]
L. fermentum
L. plantarum
L. paracasei subsp. paracasei G15Lowered circulating LPS and inflammation cytokines, such as IL-1β and IL-8, and alleviated the inflammatory status and islet β-cell dysfunction[117]
L. casei Q14
L. paracasei KBL382Increased IL-10 and growth factor-β[118]
L. plantarum ST-IIIDecrease the number of inflammatory cells[119]
L. rhamnosus GR-1TLR4[120]

Table 7.

Immune-modulating compound/mechanism by Lactobacillus.

Numerous uropathogenic bacteria can interfere with the ability of the host to eliminate pathogens by subverting cellular functions. Probiotic L. rhamnosus GR-1 affected the immunological response of E. coli challenged of bladder cells by increasing NF-kappaB activation and TNF release [120]. The urogenital probiotic L. rhamnosus GR-1 regulated NF-kappaB activation by boosting TLR4 levels on bladder cells and modifying subsequent cytokine release from urothelial cells. These lactobacilli might help pathogen identification and infection control by affecting immunological factors like TLR4, which are crucial in the fight against infections.

Immune modulation and alterations in intestinal microbiota have been associated with probiotic administration, with implications for atopic dermatitis (AD). Oral administration of L. paracasei KBL382 was shown to significantly decrease AD-related skin lesions, epidermal thickening, immunoglobulin E levels in the blood, and immune cell infiltration [118]. Immunomodulatory activity in mice of L. fermentum JDFM216 was also shown to alter gut microbiota composition thus providing the advantage of improved health through better cognition, physiological behavior, and immunity [115].

The probiotic potential of lactic acid bacteria strains isolated from Korean infant feces and Kimchi was also investigated [116]. The production of lymphocyte interferon (IFN) and cell proliferation were measured to assess the immunological modulatory activities of the strains. L. gasseri, L. fermentum, and L. plantarum strains all showed elevated IFN- levels and lymphocyte proliferation. In an in vivo model that assesses the impact of immune modifying lactobacilli on host life span using Caenorhabditis elegans as a model organism, feeding with L. plantarum CJLP133 and L. fermentum LA12 extended the average life span of the model host. Moreover, L. brevis B13–2 induced the expression of numerous cytokines (TNF-, IL-1, and IL-6) and iNOS [112].

2.8 Probiotic properties

Probiotics are live microorganisms that, when given in sufficient amounts, provide health benefits to the host. They create a favorable environment for the proper functioning of different metabolic activities in the gut, such as protein, carbohydrate, vitamin, and enzyme synthesis. Acids and the proteolytic activity of lactic acid bacteria inhibit harmful microorganisms in the intestine [123].

Colonized probiotic bacteria have a wide array of beneficial effects on the host cell, all of which are mediated via a large number of bioactive molecules. One of the mechanisms of probiotics includes competitive inhibition of the harmful bacteria by changing the pH and limiting the availability of oxygen, which leads to a less favorable environment in the intestine [123]. Probiotics also produce specific toxins with relatively narrow killing ranges, such as bacteriocins. It can also manufacture key micronutrients including vitamins, amino acids, and enzymes, boosting dietary nutrient bioavailability. Probiotics play an important role in stimulating the host immune system and enhancing the metabolic activity of carbohydrates as well [124].

Lactobacillus strains are essential components of the human and animal microbiome, and their varied impacts on host health have attracted a lot of interest [125]. Lactobacillus is one of the widely existing probiotic microorganisms. Probiotic lactobacilli have a significant and positive impact on the growth of the host, particularly in terms of enhancing body weight and size. The administration of probiotics L. casei variety rhamnosus to children with acute diarrhea showed a decrease in fecal lactoferrin and calprotectin concentrations and recovered faster as they exhibited significantly better appetite and oral intake, body weight gain, abdominal pain, bloating, and bowel movements [126]. An interleukin-22 (IL22)-secreting L. reuteri was shown to ameliorate non-alcoholic fatty liver disease [127].

2.9 Bio-converted metabolites

Dietary phytochemicals commonly occur in plant-based foods such as fruits and vegetables. These plant components with distinct bioactivities towards animal biochemistry and metabolism are being thoroughly investigated for their potential to deliver health advantages [128]. Phytochemicals often found as glyconjugates have lower bioactivity and bioavailability than their aglycone derivatives, which are smaller and less polar [129]. As a result, deglycosylation of plant glyconjugates (PGs) is recognized as a key factor in modulating their biological activity [130]. An L. acidophilus strain isolated from the human gut can activate dietary-relevant PG [131]. L. acidophilus was able to deglycosylate and externalize salicyl alcohol thus making it available for oxidation to salicylic acid by other microbial strains. This exhibits the ability of Lactobacillus to produce or mediate in the production of bio-converted metabolites (Table 8).

Lactobacillus strainBio-converted metabolitesReference
L. acidophilus NCFMSalicin[131]
L. acidophilusResveratrol[131]
L. acidophilusResveratrol[132]
L. casei
L. plantarum
L. delbrueckiiβ-maltooligosaccharides of glycitein and daidzein[133]
L. intestinalisEquol[134]
L. kimchi JB301Resveratrol[135]
L. mucosae EPI2Equol[136]
L. plantarumDaidzein[137]
L. sakei subsp. sakei
L. coryniformis
L. plantarum K2–12Daidzein and genistein[138]
L. curvatus JD0–31
Lactobacillus sp. Niu-O16S-equol[139]

Table 8.

Bio-converted metabolites produced by Lactobacillus.

Resveratrol is a phytochemical found naturally in the grape skin and seeds, wine, berries, and medicinal plants [140]. It has antioxidant, anti-inflammatory, immunomodulatory, glycemic and lipid regulating, neuroprotective, and cardiovascular protective characteristics that can help protect against a wide range of chronic diseases [141]. The bioavailability and bioactivity of resveratrol are limited due to its presence in plants in glycosidic form as piceid. To get adequate amount and activity, deglycosylation of piceid to resveratrol from plant sources is necessary. A study by Basholli-Salihu et al. [132]investigated the enzymatic ability of probiotics to transform picied to resveratrol. Cell extracts of several probiotic strains from Bifidobacterium and Lactobacillus spp., including B. infantis, Bifidobacterium bifidum, L. acidophilus, L. casei, and L. plantarum have been shown to efficiently convert piceid to resveratrol. L. acidophilus effectively converts polydatin to resveratrol as well [131].

L. mucosae EPI2 was shown to convert daidzein to equol. Daidzein is a naturally occurring isoflavone but has lesser bioactivity than its deglycosylated form equol. Equol has significantly stronger estrogenic than daidzein [136]. Bovine rumen strain Lactobacillus sp. Niu-O16, in a mixed culture with human intestinal strain Eggerthella sp. Julong 732, has also been proven to successfully synthesize S-equol from daidzein through dihydrodaidzein under anaerobic conditions [139]. High amounts of equol have been shown to efficiently lower the risk of cancer.

Advertisement

3. Conclusion

This chapter summarizes the current knowledge of the existence of diverse bioactive molecules produced by the genus lactobacillus. The diverse bioactive compounds synthesized by lactobacillus confer health and nutritional benefits to humans and animals. These compounds include bacteriocin, bioactive peptides, short-chain fatty acids, vitamins, enzymes, exopolysaccharides immune-modulating compounds, and bio-converted molecules. Collectively, the physiological function and health of the consumers are enhanced and improved by these molecules.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Zheng J, Wittouck S, Salvetti E, Franz CMAP, Harris HMB, Mattarelli P, et al. A taxonomic note on the genus lactobacillus: Description of 23 novel genera, emended description of the genus lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. International Journal of Systematic and Evolutionary Microbiology. 2020;70:2782-2858. DOI: 10.1099/ijsem.0.004107
  2. 2. EFSA Panel on Biological Hazards (BIOHAZ), Koutsoumanis K, Allende A, Alvarez-Ordóñez A, Bolton D, Bover-Cid S, et al. Update of the list of QPS-recommended biological agents intentionally added to food or feed as notified to EFSA 12: Suitability of taxonomic units notified to EFSA until march 2020. EFSA Journal. 2020;18:e06174. DOI: 10.2903/j.efsa.2020.6174
  3. 3. Tamani RJ, Goh KKT, Brennan CS. Physico-chemical properties of sourdough bread production using selected lactobacilli starter cultures. Journal of Food Quality. 2013;36:245-252. DOI: 10.1111/jfq.12037
  4. 4. Tachedijan G, Aldunate M, Bradshaw CS, Cone RA. The role of lactic acid production by probiotic lactobacillus species in vaginal health. Research in Microbiology. 2017;168:782-792. DOI: 10.1016/j.resmic.2017.04.001
  5. 5. Plengvidhya V, Breidt F, Lu Z, Fleming HP. DNA fingerprinting of lactic acid bacteria in sauerkraut fermentations. Applied and Environmental Microbiology. 2007;73:7697-7702. DOI: 10.1128/AEM.01342-07
  6. 6. Lonvaud-Funel A. Lactic acid bacteria in the quality improvement and depreciation of wine. Antonie van Leeuwenhoek International Journal of General and Molecular Microbiology. 1999;76:317-331. DOI: 10.1023/A:1002088931106
  7. 7. Song Z, Du H, Zhang Y, Xu Y. Unraveling core functional microbiota in traditional solid-state fermentation by high-throughput amplicons and metatranscriptomics sequencing. Frontiers in Microbiology. 2017;8:1294. DOI: 10.3389/fmicb.2017.01294
  8. 8. Shi Y, Singh A, Kitts D, Pratap-Singh A. Lactic acid fermentation: A novel approach to eliminate unpleasant aroma in pea protein isolates. LWT. 2021;150:111927. DOI: 10.1016/j.lwt.2021.111927
  9. 9. Jia J, Ji B, Tian L, Li M, Lu M, Ding L, et al. Mechanism study on enhanced foaming properties of individual albumen proteins by lactobacillus fermentation. Food Hydrocolloids. 2021;111:106218. DOI: 10.1016/j.foodhyd.2020.106218
  10. 10. Kemgang TS, Kapila S, Shanmugam VP, Kapila R. Cross-talk between probiotic lactobacilli and host immune system. Journal of Applied Microbiology. 2014;117:303-319. DOI: 10.1111/jam.12521
  11. 11. Gao C, Major A, Rendon D, Lugo M, Jackson V, Shi Z, et al. Histamine H2 receptor-mediated suppression of intestinal inflammation by probiotic Lactobacillus reuteri. MBio. 2015;6:e01358-e01315. DOI: 10.1128/mBio.01358-15
  12. 12. Delley M, Bruttin A, Richard M, Affolter M, Rezzonico E, Brück WM. In vitro activity of commercial probiotic lactobacillus strains against uropathogenic Escherichia coli. FEMS Microbiology Letters. 2015;362:fnv096. DOI: 10.1093/femsle/fnv096
  13. 13. Yue Y, Ye K, Lu J, Wang X, Zhang S, Liu L, et al. Probiotic strain Lactobacillus plantarum YYC-3 prevents colon cancer in mice by regulating the tumour microenvironment. Biomedicine & Pharmacotherapy. 2020;127:110159. DOI: 10.1016/j.biopha.2020.110159
  14. 14. Barbosa MS, Todorov SD, Ivanova IV, Belguesmia Y, Choiset Y, Rabesona H, et al. Characterization of a two-peptide plantaricin produced by Lactobacillus plantarum MBSa4 isolated from Brazilian salami. Food Control. 2016;60:103-112. DOI: 10.1016/j.foodcont.2015.07.029
  15. 15. Eid R, Jakee JE, Rashidy A, Asfour H, Omara S, Kandil MM, et al. Potential antimicrobial activities of probiotic lactobacillus strains isolated from raw milk. Journal of Probiotics & Health. 2016;04:138. DOI: 10.4172/2329-8901.1000138
  16. 16. Neal-McKinney JM, Lu X, Duong T, Larson CL, Call DR, Shah DH, et al. Production of organic acids by probiotic lactobacilli can be used to reduce pathogen load in poultry. PLoS One. 2012;7:e43928. DOI: 10.1371/journal.pone.0043928
  17. 17. Chen CC, Lai CC, Huang HL, Huang WY, Toh HS, Weng TC, et al. Antimicrobial activity of lactobacillus species against carbapenem-resistant Enterobacteriaceae. Frontiers in Microbiology. 2019;10:789. DOI: 10.3389/fmicb.2019.00789
  18. 18. Perez RH, Zendo T, Sonomoto K. Novel bacteriocins from lactic acid bacteria (LAB): Various structures and applications. Microbial Cell Factories. 2014;13:S3. DOI: 10.1186/1475-2859-13-S1-S3
  19. 19. Gálvez A, Abriouel H, López RL, Ben ON. Bacteriocin-based strategies for food biopreservation. International Journal of Food Microbiology. 2007;120:51-70. DOI: 10.1016/j.ijfoodmicro.2007.06.001
  20. 20. Cotter P, Hill C, Ross R. Bacteriocins: Developing innate immunity for foods. Nature Reviews. Microbiology. 2005;3:777-788. DOI: 10.1038/nrmicro1273
  21. 21. Hussein WE, Huang E, Ozturk I, Somogyi Á, Yang X, Liu B, et al. Genome-guided mass spectrometry expedited the discovery of paraplantaricin TC318, a lantibiotic produced by Lactobacillus paraplantarum strain isolated from cheese. Frontiers in Microbiology. 2020;11:1381. DOI: 10.3389/fmicb.2020.01381
  22. 22. Flórez AB, Mayo B. Genome analysis of Lactobacillus plantarum LL441 and genetic characterisation of the locus for the lantibiotic plantaricin C. Frontiers in Microbiology. 2018;9:1916. DOI: 10.3389/fmicb.2018.01916
  23. 23. Mortvedt-Abildgaard CI, Nissen-Meyer J, Jelle B, Grenov B, Skaugen M, Nes IF. Production and pH-dependent bactericidal activity of lactocin S, a lantibiotic from Lactobacillus sake L45. Applied and Environmental Microbiology. 1995;61:175-179. DOI: 10.1128/aem.61.1.175-179.1995
  24. 24. Vermeulen R, Deane S, Dicks L, Rohwer J, van Staden ADP. Manganese privation-induced transcriptional upregulation of the class IIa bacteriocin plantaricin 423 in Lactobacillus plantarum strain 423. Applied and Environmental Microbiology. 2021;87:e0097621. DOI: 10.1128/aem.00976-21
  25. 25. Wang Y, Qin Y, Xie Q, Zhang Y, Hu J, Li P. Purification and characterization of plantaricin LPL-1, a novel class IIa bacteriocin produced by Lactobacillus plantarum LPL-1 isolated from fermented fish. Frontiers in Microbiology. 2018;9:2276. DOI: 10.3389/fmicb.2018.02276
  26. 26. Jeong YJ, Moon GS. Antilisterial bacteriocin from Lactobacillus rhamnosus CJNU 0519 presenting a narrow antimicrobial spectrum. Korean Journal for Food Science of Animal Resources. 2015;35:137-142. DOI: 10.5851/kosfa.2015.35.1.137
  27. 27. Kasuga G, Tanaka M, Harada Y, Nagashima H, Yamato T, Wakimoto A, et al. Homologous expression and characterization of gassericin T and gassericin S, a novel class IIb bacteriocin produced by Lactobacillus gasseri LA327. Applied and Environmental Microbiology. 2019;85:e02815-e02818. DOI: 10.1128/AEM.02815-18
  28. 28. Garcia-Gutierrez E, O’Connor PM, Colquhoun IJ, Vior NM, Rodríguez JM, Mayer MJ, et al. Production of multiple bacteriocins, including the novel bacteriocin gassericin M, by Lactobacillus gasseri LM19, a strain isolated from human milk. Applied Microbiology and Biotechnology. 2020;104:3869-3884. DOI: 10.1007/s00253-020-10493-3
  29. 29. Acedo JZ, van Belkum MJ, Lohans CT, McKay RT, Miskolzie M, Vederas JC. Solution structure of acidocin B, a circular bacteriocin produced by Lactobacillus acidophilus M46. Applied and Environmental Microbiology. 2015;81:2910-2918. DOI: 10.1128/AEM.04265-14
  30. 30. Borrero J, Kelly E, O’Connor PM, Kelleher P, Scully C, Cotter PD, et al. Plantaricyclin A, a novel circular bacteriocin produced by Lactobacillus plantarum NI326: Purification, characterization, and heterologous production. Applied and Environmental Microbiology. 2018;84:e01801-e01817. DOI: 10.1128/AEM.01801-17
  31. 31. Chee Gor M, Golneshin A, Van TTH, Moore RJ, Smith AT. Cloning and functional expression of a food-grade circular bacteriocin, plantacyclin B21AG, in probiotic Lactobacillus plantarum WCFS1. PLoS One. 2020;15:e0232806. DOI: 10.1371/journal.pone.0232806
  32. 32. Sawa N, Koga S, Okamura K, Ishibashi N, Zendo T, Sonomoto K. Identification and characterization of novel multiple bacteriocins produced by Lactobacillus sakei D98. Journal of Applied Microbiology. 2013;115:61-69. DOI: 10.1111/jam.12226
  33. 33. O’Connor PM, O’Shea EF, Cotter PD, Hill C, Ross RP. The potency of the broad spectrum bacteriocin, bactofencin A, against staphylococci is highly dependent on primary structure, N-terminal charge and disulphide formation. Scientific Reports. 2018;8:11833. DOI: 10.1038/s41598-018-30271-6
  34. 34. Sun Z, Wang X, Zhang X, Wu H, Zou Y, Li P, et al. Class III bacteriocin Helveticin-M causes sublethal damage on target cells through impairment of cell wall and membrane. Journal of Industrial Microbiology & Biotechnology. 2018;45:213-227. DOI: 10.1007/s10295-018-2008-6
  35. 35. Rushdy AA, Gomaa EZ. Antimicrobial compounds produced by probiotic Lactobacillus brevis isolated from dairy products. Annales de Microbiologie. 2013;63:81-90. DOI: 10.1007/s13213-012-0447-2
  36. 36. Vaughan A, Eijsink VGH, Sinderen DV. Functional characterization of a composite bacteriocin locus from malt isolate Lactobacillus sakei 5. Applied and Environmental Microbiology. 2003;69:7194-7720. DOI: 10.1128/AEM.69.12.7194-7203.2003
  37. 37. Le NTT, Bach LG, Nguyen DC, Le THX, Pham KH, Nguyen DH, et al. Evaluation of factors affecting antimicrobial activity of bacteriocin from Lactobacillus plantarum microencapsulated in alginate-gelatin capsules and its application on pork meat as a bio-preservative. International Journal of Environmental Research and Public Health. 2019;16:1017. DOI: 10.3390/ijerph16061017
  38. 38. Todorov SD. Bacteriocin production by Lactobacillus plantarum AMA-K isolated from Amasi, a Zimbabwean fermented milk product and study of the adsorption of bacteriocin AMA-K to Listeria sp. Brazilian Journal of Microbiology. 2008;39:178-187. DOI: 10.1590/S1517-83822008000100035
  39. 39. Milioni C, Martínez B, Degl’Innocenti S, Turchi B, Fratini F, Cerri D, et al. A novel bacteriocin produced by Lactobacillus plantarum LpU4 as a valuable candidate for biopreservation in artisanal raw milk cheese. Dairy Science & Technology. 2015;95:479-494. DOI: 10.1007/s13594-015-0230-9
  40. 40. Zangeneh M, Khorrami S, Khaleghi M. Bacteriostatic activity and partial characterization of the bacteriocin produced by L. plantarum sp. isolated from traditional sourdough. Food Science & Nutrition. 2020;8:6023-6030. DOI: 10.1002/fsn3.1890
  41. 41. Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SS, McCulle SL, et al. Vaginal microbiome of reproductive-age women. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:4680-4687. DOI: 10.1073/pnas.1002611107
  42. 42. Dong Q, Nelson DE, Toh E, Diao L, Gao X, Fortenberry JD, et al. The microbial communities in male first catch urine are highly similar to those in paired urethral swab specimens. PLoS One. 2011;6:e19709. DOI: 10.1371/journal.pone.0019709
  43. 43. Gupta K, Stapleton AE, Hooton TM, Roberts PL, Fennell CL, Stamm WE. Inverse association of H2O2-producing lactobacilli and vaginal Escherichia coli colonization in women with recurrent urinary tract infections. The Journal of Infectious Diseases. 1998;178:446-450. DOI: 10.1086/515635
  44. 44. Gaspar C, Donders GG, Palmeira-de-Oliveira R, Queiroz JA, Tomaz C, Martinez-de-Oliveira J, et al. Bacteriocin production of the probiotic Lactobacillus acidophilus KS400. AMB Express. 2018;8:153. DOI: 10.1186/s13568-018-0679-z
  45. 45. Pessione E. Lactic acid bacteria contribution to gut microbiota complexity: Lights and shadows. Frontiers in Cellular and Infection Microbiology. 2012;2:86. DOI: 10.3389/fcimb.2012.00086
  46. 46. Pessione E, Cirrincione S. Bioactive molecules released in food by lactic acid bacteria: Encrypted peptides and biogenic amines. Frontiers in Microbiology. 2016;7:876. DOI: 10.3389/fmicb.2016.00876
  47. 47. Fernández M, Zúñiga M. Amino acid catabolic pathways of lactic acid bacteria. Critical Reviews in Microbiology. 2006;32(3):155-183. DOI: 10.1080/10408410600880643
  48. 48. Hayes M, Ross RP, Fitzgerald GF, Stanton C. Putting microbes to work: Dairy fermentation, cell factories and bioactive peptides. Part I: overview. Biotechnology Journal. 2007;2:426-434. DOI: 10.1002/biot.200600246
  49. 49. Raveschot C, Cudennec B, Coutte F, Flahaut C, Fremont M, Drider D, et al. Production of bioactive peptides by lactobacillus species: From gene to application. Frontiers in Microbiology. 2018;9:2354. DOI: 10.3389/fmicb.2018.02354
  50. 50. Yamamoto N. Antihypertensive peptides derived from food proteins. Biopolymers. 1997;43:129-134. DOI: 10.1002/(SICI)1097-0282(1997)43:2<129::AID-BIP5>3.0.CO;2-X
  51. 51. Chandra P, Vij S. Molecular characterization and identification of bioactive peptides producing Lactobacillus sps. Based on 16S rRNA gene sequencing. Food Biotechnology. 2018;32:1-14. DOI: 10.1080/08905436.2017.1413657
  52. 52. Skrzypczak K, Gustaw W, Szwajgier D, Fornal E, Waśko A. κ-Casein as a source of short-chain bioactive peptides generated by Lactobacillus helveticus. Journal of Food Science and Technology. 2017;54:3679-3688. DOI: 10.1007/s13197-017-2830-2
  53. 53. Raveschot C, Deracinois B, Bertrand E, Flahaut C, Frémont M, Drider D, et al. Integrated continuous bioprocess development for ace-inhibitory peptide production by Lactobacillus helveticus strains in membrane bioreactor. Frontiers in Bioengineering and Biotechnology. 2020;8:585815. DOI: 10.3389/fbioe.2020.585815
  54. 54. Adams C, Sawh F, Green-Johnson JM, Jones Taggart H, Strap JL. Characterization of casein-derived peptide bioactivity: Differential effects on angiotensin-converting enzyme inhibition and cytokine and nitric oxide production. Journal of Dairy Science. 2020;103:5805-5815. DOI: 10.3168/jds.2019-17976
  55. 55. Rokka T, Syväoja EL, Tuominen J, Korhonen H. Release of bioactive peptides by enzymatic proteolysis of Lactobacillus GG fermented UHT milk. Milchwissenschaft. 1997;52:675-678
  56. 56. Aguilar-Toalá JE, Santiago-López L, Peres C, Peres C, Garcia H, Vallejo-Cordoba B, et al. Assessment of multifunctional activity of bioactive peptides derived from fermented milk by specific Lactobacillus plantarum strains. Journal of Dairy Science. 2016;100:65-75. DOI: 10.3168/jds.2016-11846
  57. 57. Singh BP, Vij S. Growth and bioactive peptides production potential of Lactobacillus plantarum strain C2 in soy milk: A LC-MS/MS based revelation for peptides biofunctionality. LWT- Food Science and Technology. 2017;86:293-301. DOI: 10.1016/j.lwt.2017.08.013
  58. 58. Galli V, Mazzoli L, Luti S, Venturi M, Guerrini S, Paoli P, et al. Effect of selected strains of lactobacilli on the antioxidant and anti-inflammatory properties of sourdough. International Journal of Food Microbiology. 2018;286:55-65. DOI: 10.1016/j.ijfoodmicro.2018.07.018
  59. 59. Clare DA, Swaisgood HE. Bioactive milk peptides: A prospectus. Journal of Dairy Science. 2000;83:1187-1195. DOI: 10.3168/jds.S0022-0302(00)74983-6
  60. 60. Fan M, Guo T, Li W, Chen J, Li F, Wang C, et al. Isolation and identification of novel casein-derived bioactive peptides and potential functions in fermented casein with Lactobacillus helveticus. Food Science and Human Wellness. 2019;8:156-176. DOI: 10.1016/j.fshw.2019.03.010
  61. 61. Nakamura Y, Yamamoto N, Sakai K, Okubo A, Yamazaki S, Takano T. Purification and characterization of angiotensin I-converting enzyme inhibitors from sour milk. Journal of Dairy Science. 1995;78:777-783. DOI: 10.3168/jds.S0022-0302(95)76689-9
  62. 62. Gobbetti M, Ferranti P, Smacchi E, Goffredi F, Addeo F. Production of angiotensin-I-converting-enzyme-inhibitory peptides in fermented milks started by Lactobacillus delbrueckii subsp. bulgaricus SS1 and Lactococcus lactis subsp. cremoris FT4. Applied and Environmental Microbiology. 2000;66:3898-3904. DOI: 10.1128/AEM.66.9.3898-3904.2000
  63. 63. Topping DL, Clifton PM. Short-chain fatty acids and human colonic function: Roles of resistant starch and nonstarch polysaccharides. Physiological Reviews. 2001;81:1031-1064. DOI: 10.1152/physrev.2001.81.3.1031
  64. 64. Qian Z, Zhu H, Zhao D, Ping Y, Gao F, Lu C, et al. Probiotic Lactobacillus sp. strains inhibit growth, adhesion, biofilm formation, and gene expression of bacterial vaginosis-inducing Gardnerella vaginalis. Microorganisms. 2021;9:728. DOI: 10.3390/microorganisms9040728
  65. 65. Cremon C, Guglielmetti S, Gargari G, Taverniti V, Castellazzi AM, Valsecchi C, et al. Effect of Lactobacillus paracasei CNCM I-1572 on symptoms, gut microbiota, short chain fatty acids, and immune activation in patients with irritable bowel syndrome: A pilot randomized clinical trial. United European Gastroenterology Journal. 2018;6:604-613. DOI: 10.1177/2050640617736478
  66. 66. Kao L, Liu TH, Tsai TY, Pan TM. Beneficial effects of the commercial lactic acid bacteria product, Vigiis 101, on gastric mucosa and intestinal bacterial flora in rats. Journal of Microbiology, Immunology, and Infection. 2020;53:266-273. DOI: 10.1016/j.jmii.2018.06.002
  67. 67. Jang HJ, Lee NK, Paik HD. Lactobacillus plantarum G72 showing production of folate and short-chain fatty acids. Microbiology and Biotechnology Letters. 2021;49:18-23. DOI: 10.48022/mbl.2009.09010
  68. 68. Zhang C, Zhang Y, Liu G, Li W, Xia S, Li H, et al. Effects of soybean protein isolates and peptides on the growth and metabolism of Lactobacillus rhamnosus. Journal of Functional Foods. 2021;77:104335. DOI: 10.1016/j.jff.2020.104335
  69. 69. Malhotra MKI. Screening and in-vitro analysis of Lactobacillus reuteri strains for short chain fatty acids production, stability and therapeutic potentials in colorectal cancer. Journal of Bioequivalence & Bioavailability. 2015;07:01. DOI: 10.4172/jbb.1000212
  70. 70. Meimandipour A, Shuhaimi M, Soleimani AF, Azhar K, Hair-Bejo M, Kabeir BM, et al. Selected microbial groups and short-chain fatty acids profile in a simulated chicken cecum supplemented with two strains of lactobacillus. Poultry Science. 2010;89:470-476. DOI: 10.3382/ps.2009-00495
  71. 71. Wang G, Zhu G, Chen C, Zheng Y, Ma F, Zhao J, et al. Lactobacillus strains derived from human gut ameliorate metabolic disorders via modulation of gut microbiota composition and short-chain fatty acids metabolism. Beneficial Microbes. 2021;12:267-281. DOI: 10.3920/BM2020.0148
  72. 72. Hati S, Patel M, Mishra B, Das S. Short-chain fatty acid and vitamin production potentials of lactobacillus isolated from fermented foods of Khasi tribes, Meghalaya, India. Annales de Microbiologie. 2019;69:1191-1199. DOI: 10.1007/s13213-019-01500-8
  73. 73. Hamzehlou P, Sepahy AA, Mehrabian S, Hosseini F. Production of vitamins B3, B6 and B9 by lactobacillus isolated from traditional yogurt samples from 3 cities in Iran, winter 2016. Applied Food Biotechnology. 2018;5:105-118. DOI: 10.22037/afb.v%vi%i.18651
  74. 74. Levit R, Savoy de Giori G, de Moreno de LeBlanc A, LeBlanc JG. Effect of riboflavin-producing bacteria against chemically induced colitis in mice. Journal of Applied Microbiology. 2018;124:232-240. DOI: 10.1111/jam.13622
  75. 75. Li P, Gu Q, Yang L, Yu Y, Wang Y. Characterization of extracellular vitamin B12 producing Lactobacillus plantarum strains and assessment of the probiotic potentials. Food Chemistry. 2017;234:494-501. DOI: 10.1016/j.foodchem.2017.05.037
  76. 76. Bhushan B, Tomar SK, Chauhan A. Techno-functional differentiation of two vitamin B12 producing Lactobacillus plantarum strains: An elucidation for diverse future use. Applied Microbiology and Biotechnology. 2017;101:697-709. DOI: 10.1007/s00253-016-7903-z
  77. 77. Gu Q, Zhang C, Song D, Li P, Zhu X. Enhancing vitamin B12 content in soy-yogurt by Lactobacillus reuteri. International Journal of Food Microbiology. 2015;206:56-59. DOI: 10.1016/j.ijfoodmicro.2015.04.033
  78. 78. De Angelis M, Bottacini F, Fosso B, Kelleher P, Calasso M, Di Cagno R, et al. Lactobacillus rossiae, a vitamin B12 producer, represents a metabolically versatile species within the genus lactobacillus. PLoS One. 2014;9:e107232. DOI: 10.1371/journal.pone.0107232
  79. 79. LeBlanc JG, Levit R, Savoy de Giori G, de Moreno de LeBlanc A. Application of vitamin-producing lactic acid bacteria to treat intestinal inflammatory diseases. Applied Microbiology and Biotechnology 2020;104:3331-3337. DOI: 10.1007/s00253-020-10487-1
  80. 80. Maske BL, de Melo Pereira GV, Vale A d S, de Carvalho Neto DP, Karp SG, Viesser JA, et al. A review on enzyme-producing lactobacilli associated with the human digestive process: From metabolism to application. Enzyme and Microbial Technology. 2021;149:109836. DOI: 10.1016/j.enzmictec.2021.109836
  81. 81. Akin-Osanaiye BC, Azeez BT, Olobayotan IW. Evaluation of invertase and amylase activities of lactic acid bacteria isolated from ‘pupuru’ (an indigenous African fermented cassava staple food). Asian Journal of Biochemistry. 2019;5:1-8. DOI: 10.9734/ajrb/2019/v5i330090
  82. 82. Sieo CC, Abdullah N, Tan WS, Ho YW. In vivo study on the persistence of transformed β-glucanase-producing lactobacillus strains in the gastrointestinal tract of chickens. Journal of Animal and Feed Sciences. 2006;15:261-274. DOI: 10.22358/jafs/66898/2006
  83. 83. Zhao R, Duan F, Yang J, Xiao M, Lu L. Integrated utilization of dairy whey in probiotic β-galactosidase production and enzymatic synthesis of galacto-oligosaccharides. Catalysts. 2020;11:658. DOI: 10.3390/catal11060658
  84. 84. Rezaei A, Amirdivani S, Khosrowshahi Asl A, Malekinejad H, Zomorodi S, Hosseinmardi F. Inhibition of the angiotensin I converting enzyme (ACE) and proteolysis of non-fat probiotic yogurt. Brazilian Journal of Food Technology. 2019;22:e2018234. DOI: 10.1590/1981-6723.23418
  85. 85. Wu N, Xu W, Liu K, Xia Y, Shuangquan. Angiotensin-converting enzyme inhibitory peptides from Lactobacillus delbrueckii QS306 fermented milk. Journal of Dairy Science. 2019;102:5913-5921. DOI: 10.3168/jds.2018-15901
  86. 86. Chen Y, Li C, Xue J, Kwok LY, Yang J, Zhang H, et al. Characterization of angiotensin-converting enzyme inhibitory activity of fermented milk produced by Lactobacillus helveticus. Journal of Dairy Science. 2015;98:5113-5124. DOI: 10.3168/jds.2015-9382
  87. 87. Singh B, Katiyar D, Chauhan RS, Kharwar RK, Lall AM. Influence of UV treatment on α-galactosidase produced by Lactobacillus plantarum. Journal of Pure and Applied Microbiology. 2013;7:595-601
  88. 88. Lima M, Souza K, Pastrana L, Vieira Soares MT, Porto A. In vitro digestion as a tool for functional isolation of a probiotic potential Lactobacillus rhamnosus. Research, Society and Development. 2020;9:e3119108544. DOI: 10.33448/rsd-v9i10.8544
  89. 89. Padmavathi T, Bhargavi R, Priyanka PR, Niranjan NR, Pavitra PV. Screening of potential probiotic lactic acid bacteria and production of amylase and its partial purification. Journal, Genetic Engineering & Biotechnology. 2018;16:357-362. DOI: 10.1016/j.jgeb.2018.03.005
  90. 90. de Souza PM, de Oliveira MP. Application of microbial α-amylase in industry - A review. Brazilian Journal of Microbiology. 2010;41:850-861. DOI: 10.1590/S1517-83822010000400004
  91. 91. Singh SK, Ahmed SU, Pandey A. Metabolic engineering approaches for lactic acid production. Process Biochemistry. 2006;41:991-1000. DOI: 10.1016/j.procbio.2005.12.004
  92. 92. Amapu T, Baba A, Ado S, Abdullahi I, Hycinth D. Amylolytic potential of lactic acid bacteria isolated from wet milled cereals, cassava flour and fruits. British Microbiology Research Journal. 2016;13:1-8. DOI: 10.9734/BMRJ/2016/24509
  93. 93. Tikhomirova VE, Kost OA, Kryukova OV, Golukhova EZ, Bulaeva NI, Zholbaeva AZ, et al. ACE phenotyping in human heart. PLoS One. 2017;12:e0181976. DOI: 10.1371/journal.pone.0181976
  94. 94. Jao C, Huang S, Hsu K. Angiotensin I-converting enzyme inhibitory peptides: Inhibition mode, bioavailability, and antihypertensive effects. Biomedicine. 2012;2:130-136. DOI: 10.1016/j.biomed.2012.06.005
  95. 95. Wang J, Li C, Xue J, Yang J, Zhang Q, Zhang H, et al. Fermentation characteristics and angiotensin I-converting enzyme-inhibitory activity of Lactobacillus helveticus isolate H9 in cow milk, soy milk, and mare milk. Journal of Dairy Science. 2015;98:3655-3664. DOI: 10.3168/jds.2015-9336
  96. 96. Yousefi L, Habibi Najafi M, Edalatian Dovom M, Mortazavian A. Production of angiotensin-converting enzyme inhibitory peptides in Iranian ultrafiltered white cheese prepared with Lactobacillus brevis KX572382. International Journal of Food Science. 2020;56:2530-2538. DOI: 10.1111/ijfs.14891
  97. 97. Troelsen JT. Adult-type hypolactasia and regulation of lactase expression. Biochimica et Biophysica Acta. 2005;1723:19-32. DOI: 10.1016/j.bbagen.2005.02.003
  98. 98. Berthold-Pluta A, St. Pluta A, Garbowska M, Stasiak L. Exopolysaccharide-producing lactic acid bacteria - health-promoting properties and application in the dairy industry. Advances in Microbiology. 2019;58:191-204. DOI: 10.21307/PM-2019.58.2.191
  99. 99. Zhou K, Zeng Y, Yang M, Chen S, He L, Ao X, et al. Production, purification and structural study of an exopolysaccharide from Lactobacillus plantarum BC-25. Carbohydrate Polymers. 2016;144:205-214. DOI: 10.1016/j.carbpol.2016.02.067
  100. 100. Tang W, Han S, Zhou J, Xu Q, Dong M, Fan X, et al. Selective fermentation of Lactobacillus delbrueckii ssp. Bulgaricus SRFM-1 derived exopolysaccharide by lactobacillus and Streptococcus strains revealed prebiotic properties. Journal of Functional Foods. 2020;69:103952. DOI: 10.1016/j.jff.2020.103952
  101. 101. Rani RP, Anandharaj M, David RA. Characterization of a novel exopolysaccharide produced by Lactobacillus gasseri FR4 and demonstration of its in vitro biological properties. International Journal of Biological Macromolecules. 2018;109:772-783. DOI: 10.1016/j.ijbiomac.2017.11.062
  102. 102. Li W, Xia X, Tang W, Ji J, Rui X, Chen X, et al. Structural characterization and anticancer activity of cell-bound exopolysaccharide from Lactobacillus helveticus MB2-1. Journal of Agricultural and Food Chemistry. 2015;63:3454-3463. DOI: 10.1021/acs.jafc.5b01086
  103. 103. Riaz MSR, Mehwish HM, Fang H, Padhiar AA, Zend X, Khurshid M, et al. Characterization and anti-tumor activity of exopolysaccharide produced by Lactobacillus kefiri isolated from Chinese kefir grains. Journal of Functional Foods. 2019;63:10588. DOI: 10.1016/j.jff.2019.103588
  104. 104. London LL, Kumar AHS, Wall R, Casey PG, O’Sullivan O, Shanahan F, et al. Exopolysaccharide-producing probiotic lactobacilli reduce serum cholesterol and modify enteric microbiota in ApoE-deficient mice. The Journal of Nutrition. 2014;144:1956-1962. DOI: 10.3945/jn.114.191627
  105. 105. Abo Saif FAA, Sakr EAE. Characterization and bioactivities of exopolysaccharide produced from probiotic Lactobacillus plantarum 47FE and Lactobacillus pentosus 68FE. Bioactive Carbohydrates and Dietary Fibre. 2020;24:100231. DOI: 10.1016/j.bcdf.2020.100231
  106. 106. Tsuda H, Miyamoto T. Production of exopolysaccharide by Lactobacillus plantarum and the prebiotic activity of the exopolysaccharide. Food Science and Technology Research. 2010;16:87-92. DOI: 10.3136/fstr.16.87
  107. 107. Zhang L, Liu C, Li D, Zhao Y, Zhang X, Zeng X, et al. Antioxidant activity of an exopolysaccharide isolated from Lactobacillus plantarum C88. International Journal of Biological Macromolecules. 2013;54:270-275. DOI: 10.1016/j.ijbiomac.2012.12.037
  108. 108. Huang Z, Lin F, Zhu X, Zhang C, Jiang M, Lu Z. An exopolysaccharide from Lactobacillus plantarum H31 in pickled cabbage inhibits pancreas α-amylase and regulating metabolic markers in HepG2 cells by AMPK/PI3K/Akt pathway. International Journal of Biological Macromolecules. 2020;143:775-784. DOI: 10.1016/j.ijbiomac.2019.09.137
  109. 109. Wang X, Shao C, Liu L, Guo X, Xu Y, Lü X. Optimization, partial characterization and antioxidant activity of an exopolysaccharide from Lactobacillus plantarum KX041. International Journal of Biological Macromolecules. 2017;103:1173-1184. DOI: 10.1016/j.ijbiomac.2017.05.118
  110. 110. Tok E, Aslim B. Cholesterol removal by some lactic acid bacteria that can be used as probiotic. Microbiology and Immunology. 2010;54:257-264. DOI: 10.1111/j.1348-0421.2010.00219.x
  111. 111. Rossi G, Pengo G, Galosi L, Berardi S, Tambella AM, Attili AR, et al. Effects of the probiotic mixture Slab51® (SivoMixx®) as food supplement in healthy dogs: Evaluation of fecal microbiota, clinical parameters and immune function. Frontiers in Veterinary Science. 2020;7:613. DOI: 10.3389/fvets.2020.00613
  112. 112. Song MW, Chung Y, Kim K, Hong WS, Chang HJ, Paik H. Probiotic characteristics of Lactobacillus brevis B13-2 isolated from kimchi and investigation of antioxidant and immune-modulating abilities of its heat-killed cells. LWT. 2020;128:109452. DOI: 10.1016/j.lwt.2020.109452
  113. 113. Kook SY, Chung EC, Lee Y, Lee DW, Kim S. Isolation and characterization of five novel probiotic strains from Korean infant and children faeces. PLoS One. 2019;14:e0223913. DOI: 10.1371/journal.pone.0223913
  114. 114. Azagra-Boronat I, Tres A, Massot-Cladera M, Franch À, Castell M, Guardiola F, et al. Lactobacillus fermentum CECT5716 supplementation in rats during pregnancy and lactation impacts maternal and offspring lipid profile, immune system and microbiota. Cell. 2020;9:575. DOI: 10.3390/cells9030575
  115. 115. Park MR, Shin M, Mun D, Jeong SY, Jeong DY, Song M, et al. Probiotic Lactobacillus fermentum strain JDFM216 improves cognitive behavior and modulates immune response with gut microbiota. Scientific Reports. 2020;10:21701. DOI: 10.1038/s41598-020-77587-w
  116. 116. Lee J, Yun HS, Cho KW, Oh S, Kim SH, Chun T, et al. Evaluation of probiotic characteristics of newly isolated Lactobacillus spp.: Immune modulation and longevity. International Journal of Food Microbiology. 2011;148:80-86. DOI: 10.1016/j.ijfoodmicro.2011.05.003
  117. 117. Tian P, Li B, He C, Song W, Hou A, Tian S, et al. Antidiabetic (type 2) effects of Lactobacillus G15 and Q14 in rats through regulation of intestinal permeability and microbiota. Food & Function. 2016;7:3789-3797. DOI: 10.1039/c6fo00831c
  118. 118. Kim WK, Jang YJ, Han DH, Jeon K, Lee C, Han HS, et al. Lactobacillus paracasei KBL382 administration attenuates atopic dermatitis by modulating immune response and gut microbiota. Gut Microbes. 2020;12:1-14. DOI: 10.1080/19490976.2020.1819156
  119. 119. Zang L, Ma Y, Huang W, Ling Y, Sun L, Wang X, et al. Dietary lactobacillus plantarum ST-III alleviates the toxic effects of triclosan on zebrafish (Danio rerio) via gut microbiota modulation. Fish & Shellfish Immunology. 2019;84:1157-1169. DOI: 10.1016/j.fsi.2018.11.007
  120. 120. Karlsson M, Scherbak N, Reid G, Jass J. Lactobacillus rhamnosus GR-1 enhances NF-kappaB activation in Escherichia coli-stimulated urinary bladder cells through TLR4. BMC Microbiology. 2012;12:15. DOI: 10.1186/1471-2180-12-15
  121. 121. Nagpal R, Kumar A, Kumar M, Behare PV, Jain S, Yadav H. Probiotics, their health benefits and applications for developing healthier foods: A review. FEMS Microbiology Letters. 2012;334:1-15. DOI: 10.1111/j.1574-6968.2012.02593.x
  122. 122. Kawashima T, Ikari N, Kouchi T, Kowatari Y, Kubota Y, Shimojo N, et al. The molecular mechanism for activating IgA production by Pediococcus acidilactici K15 and the clinical impact in a randomized trial. Scientific Reports. 2018;8:5065. DOI: 10.1038/s41598-018-23404-4
  123. 123. Schepper JD, Irwin R, Kang J, Dagenais K, Lemon T, Shinouskis A, et al. Probiotics in gut-bone signaling. Advances in Experimental Medicine and Biology. 2017;1033:225-247. DOI: 10.1007/978-3-319-66653-2_11
  124. 124. George Kerry R, Patra JK, Gouda S, Park Y, Shin HS, Das G. Benefaction of probiotics for human health: A review. Journal of Food and Drug Analysis. 2018;26:927-939. DOI: 10.1016/j.jfda.2018.01.002
  125. 125. Zhang Z, Lv J, Pan L, Zhang Y. Roles and applications of probiotic lactobacillus strains. Applied Microbiology and Biotechnology. 2018;102:8135-8143. DOI: 10.1007/s00253-018-9217-9
  126. 126. Lai H, Chiu C, Kong M, Chang C, Chen C. Probiotic Lactobacillus casei: Effective for managing childhood diarrhea by altering gut microbiota and attenuating fecal inflammatory markers. Nutrients. 2019;11:1150. DOI: 10.3390/nu11051150
  127. 127. Oh J, Schueler K, Stapleton D, Alexander L, Yen C, Keller MP, et al. Secretion of recombinant Interleukin-22 by engineered Lactobacillus reuteri reduces fatty liver disease in a mouse model of diet-induced obesity. ASM Journals. 2020;5:e00183-e00120. DOI: 10.1128/msphere.00183-20
  128. 128. Dillard CJ, German JB. Phytochemicals: Nutraceuticals and human health. Journal of the Science of Food and Agriculture. 2000;80:1744-1756. DOI: 10.1002/1097-0010(20000915)80:12<1744::AID-JSFA725>3.0.CO;2-W
  129. 129. Landete JM, Curiel JA, Rodríguez H, de las Rivas B, Muñoz R. Aryl glycosidases from Lactobacillus plantarum increase antioxidant activity of phenolic compounds. Journal of Functional Foods. 2014;7:322-329. DOI: 10.1016/j.jff.2014.01.028
  130. 130. Possemiers S, Rabot S, Espín JC, Bruneau A, Philippe C, González-Sarrías A, et al. Eubacterium limosum activates isoxanthohumol from hops (Humulus lupulus L.) into the potent phytoestrogen 8-prenylnaringenin in vitro and in rat intestine. The Journal of Nutrition. 2008;138:1310-1316. DOI: 10.1093/jn/138.7.1310
  131. 131. Theilmann MC, Goh YJ, Nielsen KF, Klaenhammer TR, Barrangou R, Abou HM. Lactobacillus acidophilus metabolizes dietary plant glucosides and externalizes their bioactive phytochemicals. MBio. 2017;8:e01421-e01417. DOI: 10.1128/mBio.01421-17
  132. 132. Basholli-Salihu M, Schuster R, Mulla D, Praznik W, Viernstein H, Mueller M. Bioconversion of piceid to resveratrol by selected probiotic cell extracts. Bioprocess and Biosystems Engineering. 2016;39:1879-1885. DOI: 10.1007/s00449-016-1662-1
  133. 133. Shimoda K, Hamada H. Synthesis of β-maltooligosaccharides of glycitein and daidzein and their anti-oxidant and anti-allergic activities. Molecules. 2010;15:5153-5161. DOI: 10.3390/molecules15085153
  134. 134. Heng Y, Kim MJ, Yang HJ, Kang S, Park S. Lactobacillus intestinalisefficiently produces equol from daidzein and chungkookjang, short-term fermented soybeans. Archives of Microbiology. 2019;201:1009-1017. DOI: 10.1007/s00203-019-01665-5
  135. 135. Koa JA, Parka JY, Kwona HJ, Ryua YB, Jeonga HJ, Parka SJ, et al. Purification and functional characterization of the first stilbene glucoside-specific β-glucosidase isolated from Lactobacillus kimchi. Enzyme and Microbial Technology. 2014;67:59-66. DOI: 10.1016/j.enzmictec.2014.09.001
  136. 136. Decroos K, Vanhemmens S, Cattoir S, Boon N, Verstraete W. Isolation and characterisation of an equol-producing mixed microbial culture from a human faecal sample and its activity under gastrointestinal conditions. Archives of Microbiology. 2005;183:45-55. DOI: 10.1007/s00203-004-0747-4
  137. 137. Tsuda H, Shibata E. Bioconversion of daidzin to daidzein by lactic acid bacteria in fermented soymilk. Food Science and Technology Research. 2017;23:157-162. DOI: 10.3136/fstr.23.157
  138. 138. Lim YJ, Lim B, Kim HY, Kwon SJ, Eom SH. Deglycosylation patterns of isoflavones in soybean extracts inoculated with two enzymatically different strains of lactobacillus species. Enzyme and Microbial Technology. 2020;132:109394. DOI: 10.1016/j.enzmictec.2019.109394
  139. 139. Wang XL, Kim HJ, Kang SI, Kim SI, Hur HG. Production of phytoestrogen S-equol from daidzein in mixed culture of two anaerobic bacteria. Archives of Microbiology. 2007;187:155-160. DOI: 10.1007/s00203-006-0183-8
  140. 140. Burns J, Yokota T, Ashihara H, Lean MEJ, Crozier A. Plant foods and herbal sources of resveratrol. Journal of Agricultural and Food Chemistry. 2002;50:3337-3340. DOI: 10.1021/jf0112973
  141. 141. Meng X, Zhou J, Zhao CN, Gan RY, Li HB. Health benefits and molecular mechanisms of resveratrol: A narrative review. Food. 2020;9:340. DOI: 10.3390/foods9030340

Written By

Rodney H. Perez and Amily E. Ancuelo

Submitted: 11 November 2021 Reviewed: 18 January 2022 Published: 12 June 2022