Next Article in Journal
Feasibility of Delivering an on-Campus Food Distribution Program in a Community College Setting: A Mixed Methods Sequential Explanatory Investigation
Previous Article in Journal
An Analysis of the Effects of Psychosocial Work Environment on the Retention Intentions of Primary Care Coordinators Nursing Patients with Chronic Illness
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Allergic Diseases: A Comprehensive Review on Risk Factors, Immunological Mechanisms, Link with COVID-19, Potential Treatments, and Role of Allergen Bioinformatics

by
Fahad M. Aldakheel
1,2
1
Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
2
Prince Sattam Chair for Epidemiology and Public Health Research, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
Int. J. Environ. Res. Public Health 2021, 18(22), 12105; https://doi.org/10.3390/ijerph182212105
Submission received: 10 September 2021 / Revised: 2 November 2021 / Accepted: 16 November 2021 / Published: 18 November 2021
(This article belongs to the Section Global Health)

Abstract

:
The prevalence of allergic diseases is regarded as one of the key challenges in health worldwide. Although the precise mechanisms underlying this rapid increase in prevalence are unknown, emerging evidence suggests that genetic and environmental factors play a significant role. The immune system, microbiota, viruses, and bacteria have all been linked to the onset of allergy disorders in recent years. Avoiding allergen exposure is the best treatment option; however, steroids, antihistamines, and other symptom-relieving drugs are also used. Allergen bioinformatics encompasses both computational tools/methods and allergen-related data resources for managing, archiving, and analyzing allergological data. This study highlights allergy-promoting mechanisms, algorithms, and concepts in allergen bioinformatics, as well as major areas for future research in the field of allergology.

1. Introduction

Allergies are chronic, inflammatory disorders with aberrant immune reactions to certain environmental chemicals, which are called allergens. A number of proteins from distinct origins can behave as allergens responsible for allergic reactions from different environments [1]. Allergy symptoms range from miserable to life-threatening reaction danger. The allergic reaction develops when the immune system is exposed to a relatively harmful antigen, according to renowned allergy experts [2]. Allergies include a wide variety of reactions. Atopy, on the other hand, is a genetic predisposition to diseases in which immunoglobulin (IgE) antibodies are produced in response to even minor exposure to environmental triggers that do not bother most people. Therefore, every atopic reaction is an allergy [3]. A variety of chemical allergens (e.g., dyes, creams, fragrances in the hair, and skincare products), food allergens (e.g., genetically modified foods, tree nuts, peanuts, and eggs), and aeroallergens (e.g., dust mites, spores, pollens) can cause allergic symptoms such as skin reactions, anaphylaxis allergic rhinitis, and asthma [4,5,6].
Clemens von Pirquet, a Viennese doctor, coined the term “allergy” in 1906 after observing the hypersensitivity of his patients to typically harmless substances, such as specific foods, pollen, or dust [7,8]. Previously, allergies were used to describe a wide range of inappropriate inflammatory hyper-immune sensitive reactions. An excessive activation of specific immune system cells that induce inflammation was thought to be the root of the majority of cases. An allergic IgE mediated mechanism was later discovered to disproportionately activate specific immune system cells and to release inflammatory mediators [8]. Philip Gell and Robin Coombs proposed a new categorization system in 1963 that included immunological components and the immune process in order to identify reactions of type I to IV hypersensitivity [9,10]. Acute IgE-mediated type I hypersensitivity was referred to as “allergy” in this classification system. This features the rapid onset of hypersensitivity or allergy symptoms and develops reactions within less than 20 min after allergic exposure. Isolation and a description of the significance of IgE were the key findings of the allergy mechanism [11,12]. Kimishige Ishizaka and his colleagues originally found out in 1960 that the antibody class IgE mediates type I allergic hypersensitivity. The IgE, also known as regenic antibody or allergic antibody, was the key immunological component that might cause atopy or allergy in immune-compromised people [12].
This article will provide an overview of the risk factors and treatment for allergy disorders, as well as the allergy-promoting mechanism of viruses and bacteria and the role of allergen bioinformatics approaches in overcoming this problem.

2. Signs and Symptoms

Allergens are protein molecules found in various forms in a variety of substances. Multiple organ systems are affected by allergens, including the circulatory, cardiac, digestive, and respiratory systems. Allergens can produce edema, cutaneous reactions, hypotension, bronchoconstriction, death, and coma depending on the sensitization rate and severity. The sudden, life-threatening, and extreme hyper-immune response is known as anaphylaxis and can cause death if not treated. Numerous allergenic compounds, such as latex, can cause skin rashes and irritations, resulting in angioedema contact and dermatitis. Allergens vary in nature and source, causing moderate to severe systemic and cutaneous symptoms depending on the exposure mechanism and route of sensitization. These can be inhaled, ingested, or exposed through skin contact. Many pollen and dust allergens are microscopic airborne particles [13]. These are easily inhaled and cause symptoms in organs exposed to the allergen, such as the nose, lungs, and eyes. Mucosal irritation, a runny nose, and sneezing are the most common symptoms of allergic rhinitis (hay fever). Swelling, irritation, and redness in the eyes are all possible side effects. Allergy particles inhaled into the lungs can cause bronchial hyper responsiveness. Particular airborne allergens can be inhaled in the lungs and induce asthmatic symptoms. Coughing, bronchoconstriction, and sneezing are caused by the narrowing of the airways. The increased mucus production restricts airflow to the lungs and thickens the airways, causing a shortness of breath (bronchial hyperresponsiveness, wheezing, and dyspnea). Allergic reactions can also be triggered by the ingestion of medications and food, allergen contact, drug administration, and insect bites [14]. Food and contact allergies symptoms include hives, itchy and swollen skin, edema, vomiting, gastrointestinal discomfort, and diarrhea. Food allergies rarely result in rhinitis or respiratory (asthmatic) reactions [15]. Insect bites, drugs, medicines, and insect contact with venom lead to systemic allergic responses affecting several organs (Figure 1) [16].

3. Epidemiology

Globally, the prevalence of allergic diseases is rising rapidly in both developing and developed countries. Some studies demonstrate that, in developed countries, allergy disorders are significantly more prevalent compared to developing countries [17,18]. The prevalence of allergies or allergic diseases is determined by several factors that determine the population’s susceptibility to developing atopic conditions. The basis for allergy incidence in individuals is a mainly genetic and environmental predisposition. A total of 8–10% of the global population suffer from one or more allergic diseases, ranging from mild rhinitis to severe anaphylaxis or asthma [18]. The prevalence and causes of these allergies are summarized in Table 1.

4. Causes and Risk Factors of Allergy

Allergens, which can be found in a variety of environments, were found to be the causative agents for allergy or hypersensitivity reactions. Recognizing allergy risk factors is critical for identifying modifiable factors and individuals who may benefit from preventive measures. Risk factors can be primary, affecting the atopic disease incidence, or secondary, affecting allergic sensitization or triggering symptoms in someone already sensitized. Allergy risk factors are divided into two categories: host and environmental (Figure 1) [26].

4.1. Host Factors

Race, sex, heredity, and age are the host characteristics that influence the allergy risk, with heredity being the most important. Host factors are not currently modifiable.

4.1.1. Race

Racial disparities in the occurrence of hay fever and asthma are difficult to explain because it is difficult to separate environmental impacts and changes produced by migration from racial factors. Black people have higher levels of IgE than Caucasians [27]. There have been reports of racial differences in the outcomes of allergic diseases, with African Americans suffering disproportionately more than white people [28,29]. Fish and shellfish allergies and a higher risk of wheat allergy are significantly more prevalent in black children than white children. The risk of deadly anaphylaxis for black children is two to three times more than that of white children [30].

4.1.2. Heredity

An allergic condition can be inherited; the development of allergic diseases has a strong genetic basis. A total of 70% of homozygous twins and 40% of non-identical twins reported similar allergy problems. Allergic people have been observed to have children with comparable allergic conditions and serious symptoms [31]. Immune sensitivity is more prevalent in allergic parents than non-allergic parents. The most prevalent allergy diseases have been discovered to be hereditary. The likelihood of developing allergies seems to be genetic and associated with a malfunction in the immune system. A total of 60–80% of biparental allergic children, 30–50% of single parental allergic children, and 12% of children with no allergic family history will develop allergic disease [32,33].

4.1.3. Sex

Atopy is predominant among boys rather than girls. This sex difference can be explained by a higher sensitization rate in men compared to women for cat epithelium, grass pollen, and house dust mite. This may also explain why boys have a higher chance of developing asthma. Although this gender disparity diminishes with age, most authors report that men have a higher prevalence of specific IgE antibodies, skin test positivity, and higher total IgE levels than women. However, for several atopic disorders, at least for asthma, the prevalence of disease appears to reverse in young adulthood [34,35].

4.1.4. Age

Age affects the likelihood of allergic sensitization and atopic disease. Allergic sensitivity is high in children, especially children with an atopic history. IgE levels in infancy are at maximum and reduce quickly between 10 and 30 years of age; following that, the decrease slows down progressively [36]. Asthma is more prevalent in children below ten [37], and hay fever is most common in young adults and children [38]. Eczema is a childhood disease that begins before the age of five in 87% of adult eczema patients and has frequent remission before adulthood [39]. The prevalence of gastrointestinal allergy disease is higher in toddlers and infants [40].

4.2. Environmental Factors

Some environmental allergens are modifiable and have been the target of preventive measures. Immune modulation occurs as a result of environmental changes, which favors allergy disease development in susceptible populations. Significant environmental factors influence immune sensitization, resulting in atopy [41].

4.2.1. Passive Smoking

There is evidence that passive smoking raises serum total IgE levels and increases the risk of allergic diseases, such as allergic rhinitis, asthma, and atopic dermatitis. Passive smoking is undeniably a significant asthma risk factor [41]. The associated allergy conditions with smoking exposure have been evaluated in several studies. The results were contradictory and alternated among the protection [42,43] and negative effects of smoking [44,45] in every allergic condition; however, some studies did not discover any effects [46,47].

4.2.2. Pollution

Human, animal, and epidemiological studies all indicate that air pollutants play a significant role in the aetiology of allergic diseases, such as asthma, in terms of exacerbation and development. This involves gaseous elements, such as particulate matter (PM), ozone (O3), and nitrogen dioxide (NO2), produced by industry and automobiles [48]. NO2 can significantly raise the allergic response to inhaled allergens, according to asthmatic controlled-exposure studies [49]. O3 exposure has also been linked to an increase in worsening symptoms, respiratory infections, hospital admissions, and the need for rescue medication, peak flow rate reductions, and asthma attacks [50,51,52,53]. Gauderman et al. observed a greater chance of developing asthma for children in high O3 areas [54], while Ackermann-Liebrich et al. documented the lifelong history of physician-diagnosed asthma as a result of outdoor residential NO2 levels [55]. Many researchers have looked into the relationship between airborne traffic-related pollutants and asthma in metropolitan regions. Asthmatic children in Mexico City are highly correlated with respiratory symptoms and traffic-related air pollution [53]. Three birth cohorts’ studies were carried out by children in Germany, the Netherlands, and Sweden until the age of four or six and suggest a favorable link between medically diagnosed asthma and traffic pollution [3,56,57].

4.2.3. Dietary Habits

It is well known that, in addition to exposure to allergens and poor hygiene habits, environmental pollution and tobacco smoke, being overweight, having a low-quality diet, obesity, and a high caloric intake in adolescents and children are important environmental factors that are responsible for developing allergies [58,59]. The majority of kids with respiratory allergies, particularly asthmatics, reported poor eating behaviors, such as eating and snacking before sleep. A reduced intake of vegetables and fruit with anti-inflammatory and antioxidant properties is likely to adversely affect the prevalence/management of asthma [60]. Results from cross-sectional [61,62,63] and case-control [60,64] studies indicate that fast food consumption is significantly associated with allergic rhinitis (pollen fever) and asthma. According to Wang et al., the proportion of processed foods consumed is related to the severity and frequency of asthma attacks [65]. Diets high in vegetables, cereals, and starch have been linked to a lower risk of allergic rhino conjunctivitis [66] and asthma [67]. Evidence reveals that high-fat diets and foods are frequently associated with rhinitis [68,69], asthma [63,67,70], respiratory health [71], and allergies, though some studies have not found these associations [72].

4.2.4. Infections

Bronchial asthma and allergic sensitization are linked to respiratory infections, particularly viral infections. Childhood allergic sensitization, followed by wheezing respiratory tract illnesses caused by respiratory pathogen infections, appears to be reproducible and consistent among the various environmental risk factors implicated in the development of childhood asthma [73,74,75]. Preschool wheezing illnesses caused by both viral [76,77,78] and bacterial [79,80,81] pathogens are also linked to an increased asthma risk and recurrent wheezing. The role of bacterial and viral infections in the development of allergic diseases is discussed later.

5. Mechanism of Allergy and Immune System

The role of the immune system is to protect the body against invading pathogens causing different diseases. When the immune system misidentifies a harmless foreign antigen as a pathogen, an allergic reaction occurs [82]. To protect the organism against exaggerated stimulation signals from harmless antigens, such as environmental and self-antigens, the immune system must be closely monitored. In genetically predisposed individuals, an imbalance in the immune system’s regulatory mechanisms may lead to allergic diseases or autoimmune disorders, depending on the nature of the antigen [83,84].
During an allergic reaction, the immune system must detect pathogenic stimuli and generate a robust immune response. Specific antigen sensitization is required: naive T and B cells identify specific sections of antigens, which are termed epitopes. First, specific MHC (major histocompatibility complex) class II antigens synthesized on the antigen-presenting cells (APC) surface detect allergens and deliver them to naive T lymphocytes. T cell activation causes T helper type 2 (TH2) cells to proliferate and differentiate. Interleukin IL-5, IL-4, and IL-13 and innate (ILC-2) lymphoid cells that can maintain and enhance local TH2 inflammation caused by the secretion of TH2 cytokines (IL-13 and IL-5) are the primary cytokines responsible for the allergic response [85]. These ILs act on B cells, causing them to switch to the Ig class E (IgE). Allergen-specific IgE antibodies bind to high-affinity IgE receptors (FcRI) on basophils and mast cells. Repeated exposure to the allergen causes FcRI-bound IgE to crosslink, boosting the release of other mediators and histamine that generate allergic disease symptoms. Allergen-specific cells are enlarged and reactivated locally after 6–12 h of allergen exposure, culminating in the late phase of an allergic reaction. Effector cells (basophils, mast cells, and eosinophils in particular) release cytokines and inflammatory mediators, prolonging the proinflammatory response (Figure 2). The symptoms of allergic disorders are caused by this phase, and persistent allergen exposure causes the disease to become chronic [83,86].
Specific antigen sensitization is required for allergic diseases development. Inflammatory cytokines (IL-13, IL-4, and IL-5) are produced as a result of cell expansion and differentiation to TH2 cell subtypes. They regulate the activation and recruitment of pro-inflammatory cells (mast cells and eosinophils) in mucosal target organs, as well as the class switching of IgE in B cells. Allergy symptoms and inflammation are triggered by these activations [87].

6. Allergy and Microbiota

The microbiota (intestinal microflora) are a collection of microorganisms, primarily bacteria, that form a complex ecosystem in the human digestive tract. Microbiota are influenced by a wide range of environmental and nutritional factors, and play a complex role in allergic diseases. According to a recent study, gut microbiota has a substantial impact on immune system development. The gut microbiota plays a significant role in the formation of immune system organs and help to identify host immune response patterns. According to research on the relationship between gut microbiota and immune diseases, modifications in commensal bacteria can trigger immune system changes that affect immune system maturation, oral tolerance development, and host metabolism regulation [88,89]. Due to the fact that the immune system is regulated by the normal intestinal ecosystem, the risk of allergy or atopy is likely to increase as the dysbiosis of the gastrointestinal tract worsens. Dysbiosis is described as a disruption in gut homeostasis caused by a change in the function and composition of the microbiota [90]. Numerous studies suggest that dysbiosis intestinal, or quantitative and qualitative abnormalities in the microflora composition, may be a factor in the pathogenesis of a variety of disorders, including inflammatory bowel disease, necrotizing enterocolitis in newborns (NEC), celiac disease, irritable bowel syndrome, atopic dermatitis, allergic disorders, cancer, depression, and others [91,92,93]. Many atopy and allergy patients have altered microbiota [94], as evidenced primarily by stool microbiota analysis [95,96,97]. Dysbiosis has also been found in the lower and upper respiratory tract microbiota of asthma patients [98], as well as the skin microbiota of atopic dermatitis patients [99,100], and in the gastrointestinal tract of food allergy sufferers [101,102].
There is mounting evidence that dysbiosis precedes the onset of allergic symptoms. A lack of specific bacterial species from the gut microbiota among infants aged 1–3 months was linked to a higher risk of developing a recurrent wheeze, asthma, or atopy later in life, according to birth cohort studies [103,104]. Such changes were associated with reduced levels or a lack of anti-inflammatory polyunsaturated fatty acids [104]. Proteobacteria, especially Haemophilus spp., are more prevalent in asthmatic adults’ lungs than in healthy controls, who have a higher proportion of Bacteroidetes. Furthermore, asthmatic children have a higher abundance of Proteobacteria than healthy controls [105]. Intestinal dysbiosis in egg-allergic children was marked by an increase in the Lachnospiraceae and Streptococcaceae genera, as well as a decrease in the Leuconostocaceae families, when compared to non-food-allergic controls [106]. Ege et al. compared the microbial data of 489 school-aged children from rural and urban areas in Germany and found a number of bacteria, including Lactobacillus, Staphylococcus, and Acinetobacter, that were inversely related to asthma and hay fever [107]. When children with allergic airway diseases were compared to children from similar surrounding environments, such as both from urban areas, a mild reduction in microbiota diversity was observed, and microorganisms from the phylum Firmicutes were significantly less expressed than in healthy children [108]. A similar pattern was observed among Swedish children. At infancy, children with asthma have a lower diversity of gut microbes than children without asthma [109].
Breast milk provides immune factors, such as IgA antibodies, that protect against a variety of health problems in infancy, including obesity and being overweight, necrotizing enterocolitis, diabetes, infections, and allergic disease [110,111], as well as reducing the risk of diseases later in life [112]. Breastfeeding, on the other hand, has been the subject of debate in the literature regarding its ability to protect children from developing asthma and allergic disease [113]. Epidemiological studies in the debate over whether breastfeeding can protect against allergic disease and asthma in early childhood provide contradictory results [112]. While breastfeeding is advised for all infants, regardless of allergic history [114], with protective effects of breastfeeding on asthma reported in young children [115,116], other studies of children at low [117] or high risk [118,119], or adults [120], have found no protective effects.
Dysbiosis may start even earlier, according to recent research, as meconium from at-risk neonates displays an altered microbiota-derived metabolome and delayed gut microbial diversity, characterized principally by a lack of anti-inflammatory fecal lipids [121]. This dysbiosis is significantly linked to parental sickness, implying that maternal health during pregnancy may have an impact on the vertical transmission of microbes that affect early microbiota development. Changes in the intestinal microbiota composition may induce food allergy resistance or vulnerability via a microbially responsive FOXP3+ RORt + Treg cell subset, which is known to be critical for food tolerance maintenance [122].
Mouse studies, particularly experiments with germ-free mice, provide experimental evidence for the association of microbiomes with allergy development. Germ-free mice have an adaptive immune response profile reprogrammed. They are especially predisposed to TH2 cell development [123,124]. Germ-free mice may be reconstructed by specific microbial strains and allergy protection is induced by Clostridia and other allergy-protective-related species through IgA production, Treg cell induction, and other immunologic effects [125,126].
It was recently demonstrated that germ-free mice colonized with healthy infants commensals, but not colonized with commensals from cow’s milk allergic infants, were protected against anaphylactic responses to cow’s milk allergens. The Clostridia member, Anaerostipes caccae, was further found in this model as protection against allergic response to food [127]. In another food allergy model, colonization with seven species of Bacteroidales consortium or a Clostridiales consortium suppressed food allergy in the mouse model [128]. Further studies revealed that commensals activated the MyD88–RORt pathway, resulting in the development of Treg cells [128]. These significant experiments aid in a better understanding of the functional significance of dysbiosis in patients with food allergies [129].
Hence, we can conclude that environmental and dietary changes cause dysbiosis in the gut, skin, and/or lung microbiome, resulting in quantitative and qualitative alterations in the microbiota that directly alter immunological pathways implicated in allergic disorders prevention. More research is needed, however, to determine the cause-and-effect link between the microbiota and asthma/allergy clinical phenotypes.

7. Viral Infections in Allergy

Viral infections can have a variety of opposing effects on allergy and asthma development; depending on the circumstances, viruses can either protect against or trigger allergic disorders. During the first year of life, the immune system and respiratory tract mature quickly, and postnatal lung development is influenced by and affects viral infection responses. The type of virus, age, intensity, timing, and location of the infection, as well as interactions with pollutants or allergens, have all been linked to allergic diseases development, particularly asthma, regarding viral infections [130]. By binding to certain receptors on the airway epithelial cells surface, viruses trigger antiviral and inflammatory responses, resulting in the innate immune responses activation, the recruitment of mononuclear and neutrophil cells to the area, and the release of mediators, such as chemokines and cytokines [130,131]. Such events can alter immunological and epithelial responses to a hyperactive state [132].
Viral respiratory tract infections and allergens can interact in a variety of ways, including through a flawed epithelial barrier function. Viral respiratory tract infections are related to an impaired innate immunity, suppressed antioxidant properties, and disrupted tight junctions, which may result in a hypersensitivity to allergens and infections [133]. Asthma development has also been linked to viral infections. Asthma onset in childhood and asthma exacerbations in adults and older children are linked to viral respiratory tract infections. The respiratory viruses linked to asthma include those that cause influenza-like illnesses, the common cold, and bronchiolitis, as well as wheeze in children. In school-aged children and adults, respiratory viruses represent approximately 85% of exacerbations of asthma [134,135]. The respiratory syncytial virus (RSV), human rhinoviruses, and influenza viruses are among the viruses linked to asthma exacerbation. Not only can RSV infections induce asthma but, according to an epidemiological study, they can also lead to allergic sensitization and asthma development [136]. Parainfluenza viruses, coronaviruses, adenoviruses, and the newly discovered bocaviruses and metapneumoviruses are also involved, but they are less common [137,138]. Although it is unclear how viruses influence asthma onset, various studies have been carried out about the host response to respiratory viruses and how viruses can induce the host response or how subsequent allergen exposure and sensitization affect the host response.

7.1. Allergy Promoting Mechanism of Viruses

Although the mechanisms underlying the association of asthma and viral respiratory tract infection are not completely understood, recent reports indicate that epithelial cell viral infection could produce cytokines, such as IL-33 and IL-25, that interact with allergic inflammation, inducing both antigen-specific and innate TH2 cell–related pathways, and resulting in mucin production, increased TH2 related inflammation, enhanced IL-13, IL-4, and IL-5, and eosinophilia [139,140]. In patients with atopic asthma, the TH2 cytokines (IL-13, IL-5, and IL-4,) are well known as effector molecules [134]. The effects of viruses and allergens on immune and airway epithelial cells, as well as the elicitation of TH2 responses, are summarized in Figure 3.
Viral infection disrupts the epithelial barrier, resulting in the thymic stromal lymphopoietin and pro-TH2 cytokines IL-25 and IL-33. These cytokines act on TH2 cells, DCs, and ILC2s, causing the TH2 cytokines IL-13, IL-5, and IL-4 to be produced. These cytokines are important in asthma: IL-13 and IL-4 promote antibody class switching to IgE in B cells, IL-13 can also act on smooth airway muscle cells, causing bronchoconstriction and aiding in the remodeling of airways, and IL-5 stimulates the production of eosinophil. IL-4, IL-13, and virus actions on airway epithelial cells can elicit eotaxins, which attract eosinophils, as well as activation-regulated chemokine (TARC), the chemokines macrophage-derived chemokine (MDC), and thymus, which attract TH2 cells into the airway. IgE cross-linkage with allergens in mast cells releases leukotrienes, histamine, and the prostaglandins PGE2 and PGD2, which promotes bronchoconstriction. PGD2 activates ILC2s, TH2 cells, and basophils by binding to CRTH2, a chemoattractant receptor-homologous molecule expressed on TH2 cells. Oxidative stress can also be caused by viruses, and the formation of pathogen-associated molecular patterns (PAMP) and damage-associated molecular patterns (DAMP) can lead to pro-inflammatory cytokines, such as IL-6, TNF, and IL-1a/b. Propagative cytokines are generated. This usually results in macrophage activation and neutrophilic inflammation. Allergen-induced IL-1a can also stimulate the pro-TH2 response, resulting in ILC2 activation and IL-33 production [141].

7.2. Allergy and COVID-19

COVID-19, caused by SARS-CoV-2 [142,143], shares many symptoms with allergic diseases, such as coughing, olfactory, shortness of breath, nasal congestion, and taste dysfunction [144,145]. Some allergic disorders, such as chronic rhinosinusitis with nasal polyps (CRSwNP), allergic rhinitis, and asthma, can simulate COVID-19 symptoms: asthmatic patients experience cough and dyspnea, whereas allergic rhinitis and CRSwNP patients experience runny noses and headaches [146]. Multiple pathophysiological processes indicate that allergies may increase the risk of SARS-CoV-2 infection [141]. The respiratory virus initiates a local inflammatory cascade, resulting in cytokine production, which can aggravate asthma and allergy symptoms [139]. Besides, allergic patients have impaired innate interferon secretion, increasing their susceptibility to respiratory viral infections [141]. Besides, this pandemic began in the spring, when seasonal allergy sufferers are most likely to experience some of the same symptoms [147]. Chronic airways diseases and the COVID-19 pandemic are both associated with anxiety, which should be considered when interpreting subjective symptoms of both conditions. COVID-19 patients have been documented to have skin symptoms and signs of eczema and urticaria that are similar to acute urticaria or medication reactions, creating a diagnostic difficulty for allergists and dermatologists. For this reason, it is important to pay attention to COVID-19-specific symptoms, such as mainly fever, as well as excessive fatigue and a diminished sense of taste or smell, in order to make an accurate diagnosis [146,147].
Adults and children with allergies are more prone to have physical and mental health concerns during the COVID-19 pandemic. COVID-19 and allergens are independently associated with mental health problems [148,149,150]. Gonzalez-Diaz, et al. reported that patients with allergic diseases were more affected psychologically by the COVID-19 quarantine than those without allergies, as allergic individuals had a higher risk of depression symptoms [151]. Allergic patients were more likely to engage in various COVID-19 preventive measures, including maintaining a six feet social distance, avoiding crowded or public places, wearing a face mask, postponing or canceling activities, avoiding contact with high-risk people, and sanitizing or washing their hands [152]. A greater adherence to COVID-19 preventative activities showed the significant impact of the pandemic on the mental health of this group, because social isolation can cause hopelessness and depression [153]. Interactions between COVID-19 and allergy-related inflammatory psychiatric disorders, such as anxiety, post-traumatic stress disorder (PTSD), and depression, have been reported [154]. The stress of the COVID-19 pandemic, therefore, may increase the psychiatric reaction in those who have preexisting allergic conditions.
The COVID-19 epidemic has been a burden for allergy professionals. Since COVID-19 shares similar allergy disease symptoms, a pandemic may cause a problem in prioritizing allergic people, face-to-face assessment, and further concerns about the potential diagnostics of COVID-19. Face-to-face and hospital visits should be kept to a minimum for allergic disease patients, and more attention and promotion should be given to social distancing, hand disinfection, patient consultation adaptations, and sufficient PPE for health care employees. Teleconsultation for allergic patients during COVID-19 is very promising, and telemedicine platforms can provide a trustworthy service [155,156].

8. Bacterial Infections in Allergy

Bacteria play a dual role in allergies. They mainly concern protection, although certain species of bacteria stimulate allergic inflammation. Bacterial exposure has long been linked to allergy prevention. For example, mycobacteria are potent inducers of Th1 responses, notably IFN-release, which counteract type 2 inflammation and elicit regulatory T cell (Treg) responses, the primary anti-allergic immunological mechanism [87]. Mycobacterium tuberculosis infection and vaccination with other mycobacteria or Bacillus Calmette-Guérin reduce allergy prevalence in animals and humans [157,158,159]. Furthermore, there is a wealth of evidence suggesting bacterial compounds influence the innate immune system. TLR4 and other innate pattern recognition receptors play an important role in anti-allergenic effects [160].
In recent decades, the hygiene theory has been bolstered by the discovery of a considerable decline in infectious diseases associated with a sharp increase in the frequency of allergy: “The decline in the incidence of infectious diseases in industrialized countries throughout the past three decades is the major explanation for the increased prevalence of allergy diseases in those countries” [161]. As the role of commensal microbiota in immune regulation and inflammatory homeostasis became more apparent, this hypothesis was later modified. Early exposure to innocuous endogenous and exogenous microorganisms reduces allergy risk. In general, the alterations in the microbiome might affect allergy manifestations, both in terms of their diversity and abundance [162,163,164]. Due to this observation, the ability of specific commensal gut microflora species (probiotic strains) to promote immunological tolerance, notably lactic acid bacteria, including Bifidobacteria or Lactobacillus, is currently being examined. Several reports exist that detail the significance of these strains in allergy disorders prevention [165,166,167].
On the other hand, epidemiological evidence suggests that infection or colonization with specific bacterial species might cause or worsen allergies [163,168]. Bacteria, for example, can aggravate asthma symptoms on their own or in combination with viruses, such as the respiratory syncytial virus or human rhinovirus [169,170]. Studies in the 1970s and 1980s showed that bacterial colonization was linked with allergy disorders. Atypical bacteria, such as Mycoplasma pneumonia, Chlamydia pneumoniae, and Chlamydia trachomatis, have been associated with asthma exacerbations, lung remodeling, and an increased incidence of wheezing episodes. These pathogens have also been found in nasal washes, sera, and bronchoalveolar lavage fluid (BAL) from asthmatic patients [171,172,173,174]. Infection or colonization with Streptococcus pneumoniae, Staphylococcus aureus, Moraxella catharralis, and Haemophilus influenzae, among the common human respiratory tract bacterial inhabitants, has been linked to recurrent wheezing in children, obstructive pulmonary disease, and the onset and exacerbation of asthma, [169,170,175,176]. Furthermore, the asymptomatic M. catarrhalis or S. pneumoniae colonization of newborns is linked to asthma development and a recurrent wheeze later in life [175].

Allergy Promoting Mechanism of Bacteria

Bacteria have been found to exhibit a variety of pro-allergenic activities (non-antigen-specific and antigen-specific). Airway epithelial cells can be infected by bacteria, causing cell death, inflammation, and the breakdown of the epithelial barrier. Pore-forming toxins, such as bacterial proteases and S. aureus toxins (Hla), also play a role in epithelial barrier breakdown. Microbial invasion is facilitated by an increased epithelial permeability, which exposes the immune system to allergens and environmental pollutants [177].
Some bacteria can cause histamine release from mast cells and human basophil leukocytes via independent or IgE-dependent mechanisms [178,179]. Several studies have shown that bacteria can cause both naive T cells to differentiate into TH2 or TH17 cells and the release of TH2 cytokines [178,180,181].
The induction of cytokines type 2 is likely to result in a switching of the Ig class to IgE. IgE antibodies directed against C. pneumoniae, C. trachomatis, H. influenzae, S. aureus, M. pneumoniae, M. catharralis, or S. pneumonia have been described [171,175,181,182]. Despite the fact that antibacterial IgE can be measured, there is evidence of allergy protection from exposure to S. pneumonia or H. influenzae. Specific IgE antibodies were found to be inversely linked to asthma risk in teens in diverse proteins of these microbial species. Furthermore, they emphasize the significance of the mechanistic and epidemiological validation of allergen prediction [183].
House dust mites (HDM) have recently been shown to be antigen carriers for bacteria colonizing the respiratory tract, skin, or gut, such as E. coli or S. aureus. Hence, HDM could cause or aid bacterial antigen sensitization. This may help to explain why the IgE response to bacterial antigens is so common in skin and respiratory allergy symptoms [184].
Hence, bacteria exert control over general allergy-inducing pathways and may become targets for type 2 immune responses defined by IgE antibodies and specific TH2 cells [183]. Figure 4 depicts an overview of bacteria’s allergy-promoting processes.
Bacteria have described a number of pro allergenic pathways. Bacterial toxins and proteases break down the epithelial barrier, allowing for the entry of conventional allergens and microbial invasion. This causes the release of potent immune mediators (TSLP, IL-33, and IL-25) and local inflammation (a). This pathway helps to recruit and differentiate naive T-cells into effector T-cells (TH17 and TH2), which leads to the release of pro-allergenic Th2 cytokines. Type 2 cytokine secretion is also elicited by tissue-resident ILC2s (b). B cells differentiate into IgE-secreting plasma cells after undergoing an Ig class switch (c). IgE promotes the activation and recruitment of basophils, mast cells, and eosinophils (d). Bacterial components can potentially cause the IgE-independent degranulation of these effector cells, increasing allergic inflammation (e) [183].

9. Treatments

Advances in allergy research have made a significant impact on the treatment of moderate to severe allergic disorders. Numerous treatments for different symptoms of allergic disorders, as well as several drugs that effectively control and treat atopic conditions, are available. Epinephrine shots for anaphylaxis are available and can be carried with the patient, while anti-inflammatory and antihistamine medicines are commonly administered to relieve symptoms in others [185,186].
The treatment of allergic diseases in children follows a similar pattern to that of adults. Treatment options include allergen avoidance through environmental control, pharmacotherapy, and immunotherapy. The main goal of treatment is to control symptoms without affecting the child’s functioning. The second, but equally important, goal is to prevent the development of the sequelae of allergic diseases. Currently, the best approach for a child at a high risk of developing allergies is to implement dietary and environmental control measures early in order to reduce sensitization, and to acknowledge and treat the signs and symptoms of allergic disease as they emerge [187,188,189].
Various methods of diagnosis have been created based on allergen sources, and therapy strategies based on diagnostic methods have been developed to address allergic reactions concerns. Some of the treatments utilized for allergic diseases are the following.

9.1. Allergen Avoidance

The primary focus of allergy treatment should always be the strict avoidance of specific allergens that cause allergic disease. The greatest and best guideline for reducing allergy reactions in sensitive people is to avoid allergen exposure. Food allergies and some stinging insect allergies are treated primarily through avoidance, which can be quite helpful if patients are well trained about preventive measures. However, it is impossible to avoid certain allergens that travel through the air and are easily inhaled without control or notice. Avoidance is impossible in these circumstances, and additional therapeutic procedures are necessary to overcome difficulties [16,190].

9.2. Pharmacotherapy

Pharmacotherapy can relieve allergen-induced symptoms when allergen prevention and tracking are impossible and allergy exposure is inevitable. Many drugs are developed that are antagonistic to and block the actions of allergic mediators. Anti-leukotrienes and antihistamines are two common drug targets that prevent the onset of allergic symptoms and inhibit the action of inflammatory mediators [185,186]. The FDA has approved adrenaline (epinephrine), antihistamines, glucocorticosteroids, and theophylline, which primarily act as anti-inflammatory molecules. Decongestants, mast cell stabilizers, and eosinophil chemotoxins, along with anti-leukotrienes, such as zafirlukast (Accolate) or montelukast (Singulair), are commonly used as drugs to monitor and prevent chronic and acute allergic diseases.

9.3. Immunotherapy

Allergen-specific immunotherapy entails administering an increasing dose of allergens to a patient over time to ensure immunological and clinical tolerance. Allergen injection immunotherapy induces T cell tolerance through a variety of methods, including alteration in secreted cytokines, decreased allergen-induced proliferation, stimulation of apoptosis, and T regulatory cells production. This results in the reduction of inflammatory mediators and cells in the affected tissues, production of blocking antibodies, and suppression of IgE [191]. This sort of immune therapy has been demonstrated to be effective in studies, and long-term use has indicated that immunotherapy can help to avoid the development of atopy. The intravenous administration of monoclonal anti-IgE antibodies is the second type of immunotherapy. These attach to both B-cell-associated and free IgE, signaling and killing them [192]. Sublingual immunotherapy is a third type of therapy that is given orally and is based on oral immune tolerance to non-pathogens, such as resident bacteria and foods. Allergy shot therapy may become the most effective allergy treatment method in the future. Close supervision and a long-term commitment are required in this therapy for successful individual treatment [193].

9.4. Ineffective and Unproven Treatments

An enzyme potentiated desensitization (EPD) experimental treatment has been tested in some recent investigations, but no encouraging outcomes have been found. The same method is currently used in many hypoallergenic food preparations. The treatment approach, however, was not convincing, and was not acknowledged as effective. EPD uses allergen dilutions with beta-glucuronidase enzymes to polarize T-regulatory lymphocytes and to change the allergen nature, which down-regulates IgE induction, favors desensitization, and prevents allergic reactions [194].

10. Role of Bioinformatics in Allergic Diseases Management

Allergy research has progressed quickly in recent years [195]. Recent advances in proteomics, analytical methods, and genomics have resulted in massive amounts of allergen-related data. The pathophysiology of many allergy conditions based on epidemiologic, experimental, and clinical information for allergic reactions can be related to this data. A continuous data increase requires effective archival, data management, and data analysis. In the modern era, bioinformatics applications are used to predict allergens and their allergenicity. Bioinformatics complements wet-lab research by providing tools for managing this avalanche of data. Despite the fact that a large amount of biological data is difficult to manage, specific tools and databases are available to handle data. Several tools, databases, and servers contain a wide range of information about allergens and other potential side effects. The goal of allergy-related databases is to make data retrieval, collection, and analysis easier. Furthermore, bioinformatics techniques can be used to organize allergens and to identify areas that may account for common IgE binding patterns and cross-reactivity [196]. These findings can be used to help allergy sufferers choose the best treatment options. Hence, the discipline of allergy bioinformatics has emerged, which includes allergen-specific resources/databases, as well as computational tools/methods [197]. Many research papers on allergen bioinformatics and immunoinformatics have been published by various groups of researchers [198,199]. For example, Zhang et al. identified key genes and Le Chen et al. identified hub genes in a murine model in allergic rhinitis by bioinformatics analysis [200,201]. Deocaris et al. used bioinformatics analysis in the detection of nascent allergens in GMO and conventional rice [202]. L’Hocine et.al identified allergens from Canadian mustard varieties of Brassica juncea and Sinapis alba [203]. Chenbei et.al performed a bioinformatics analysis of the dataset to identify pathways and potential different expressed genes (DEGs) related to childhood allergic asthma [204].
Allergen bioinformatics deals with tools/algorithms for allergenicity/allergen prediction, allergenic cross-reactivity prediction, allergen databases, and allergen epitope prediction [197].

10.1. Allergen Cross-Reactivity Prediction

Cross-reactivity has a major role in clinical and immunological allergic reactions. The prediction of cross-reactivity in allergy was therefore considered to be significant [205]. In the majority of cases, the prediction of allergenicity is associated with the allergens cross-reactivity prediction. This is due to the fact that the antigenic determinants that cause allergen cross-reactivity are also responsible for allergenicity [206]. Hence, many of the algorithms/tools designed to predict allergen/allergenicity can also predict cross-reactivity. The criteria set by FAO/WHO experts aid in the allergen cross-reactivity identification [207]. Stadler and Stadler [208] proposed a sequence-based approach and claimed that a motif-based strategy outperforms the WHO/FAO guidelines for cross-reactivity calculations. AllerTool [209] is a cross-reactivity webserver based on the WHO/FAO guidelines and amino acid sequence. It also depicts published and projected allergen cross-reactivity patterns graphically. A sequence-based technique for determining the allergen cross-reactivity is included in SDAP [210], a specialized allergen database. AllerHunter [211] is a web server based on SVM that efficiently analyzes allergen cross-reactivity in proteins. A recently developed algorithm for an allergenicity prediction based on a fuzzy inference system can also predict allergen cross-reactivity [212].

10.2. Allergen Databases

Significant technological advances in the fields of proteomics and genomics, as well as considerable improvements in analytical methods, have occurred in recent years. As a result, significant progress has been made in allergy research. Hence, the number of identified protein allergens has been steadily increasing in recent years [213,214]. As a result of the constant accumulation of allergen-related clinical and molecular data, efficient data storage and management has become critical. Allergy databases are therefore very essential resources for fundamental allergy research because they are used to archive available knowledge about allergens [215]. Table 2 provides a summary of allergen-specific databases.

10.3. Allergen/Allergenicity Computational Prediction

Allergens are primarily proteins found in pollens, food, and other biological entities in the environment. As a result of the health risks associated with allergic reactions to these proteins, it has become necessary to evaluate their potential allergenicity. Food processing and genetic engineering methods have been frequently used in recent years to modify the existing or new proteins. Analyzing the allergenicity of such products/proteins and biopharmaceuticals is critical to avoid the allergenic molecule transfer. The most common method for assessing allergenicity is computational prediction or evaluation, and a range of bioinformatics methods/tools have been effectively used for this purpose [223]. Most of these strategies are based on amino acid sequences and their different properties, with only a few approaches based on structural information [224]. The list of computational servers/tools available for the allergenicity/allergen prediction is shown in Table 3.

10.4. Allergens Epitopes Computational Prediction

Epitopes are distinguishing amino acid residuals on antigens and are significant predictors of immune responses. The identification of epitopes is considered to be a key step in the creation of effective multi subunit vaccines, as well as efficient therapeutic and allergy diagnostic procedures. IgE binding epitopes, also known as B cell epitopes, are proteins that recognize IgE binding sites in allergens. They play a significant role in the interaction between the allergen and IgE antibody. IgE-binding epitopes have distinct characteristics that distinguish the antibody epitope from other epitopes. Complex allergens and antibodies are widely used in allergen immunotherapy and aid in the understanding of allergy phenomena. There are a large number of epitopes in databases that can be used as a template for novel epitope predictions [229].
The antigen contains T cell epitopes; it is also known as the antigenic determinant that interacts with the T cell through T cell receptors. Allergen T cell epitopes have been discovered to play an important role in allergic reactions. As a result, the identification of allergic disorders would be cured by developing new epitope-based immunotherapy, which would allow for the production of effective vaccines [229].
Although experimental methods have proven to be effective in discovering epitopes, their utility is restricted due to their high cost and time requirements, as well as their incapacity to deal with large-scale epitope elucidation. Computational approaches are therefore deemed highly useful because they are cost effective and time efficient. A wide range of highly effective algorithms and methods for the prediction of epitopes have been developed throughout the years. Both T cell and B cell epitopes, including discontinuous (conformational) and sequential (linear) epitopes, are predicted using these approaches [230,231]. Table 4 lists some of the most popular tools and servers for B cell and T cell epitopes prediction.

11. Conclusions and Future Perspectives

Allergies are a severe problem that affects millions of individuals throughout the world. It may be difficult to avoid offending allergen exposure if the causative allergen is rare or unknown. Allergic patients can, however, minimize symptoms by avoiding allergen exposure. Currently, available diagnosis and treatment methods aim to alleviate symptoms; however, medication would not provide long-term relief from allergic disorders. Researchers are conducting new studies and investigations to find solutions for allergy treatment. Advances in analytical, proteomic, and genomic approaches have resulted in a massive amount of data concerning allergens and allergies. In allergen bioinformatics, analyzing and archiving these data poses a significant challenge. The tools and resources of bioinformatics play a crucial role in overcoming this challenge. With the ever-increasing volume of data, it is critical to focus on the development of resource/databases that will integrate and provide quick access to information from literature and other sources. An analysis of such data can be used to have a clear understanding of allergic reactions. Allergen structural properties influence allergenicity significantly; this knowledge is used to develop effective methods for predicting allergen cross-reactivity and allergenicity/allergen. Recent epitope prediction advancements have focused on antibody-specific epitope prediction methods. The use of these techniques for IgE-binding epitopes predictions will be critical in the development of better and more efficient strategies for allergic disease treatment and diagnosis. Allergen immunotherapy (AIT), a treatment approach based on allergens, has been regarded as a prototype of personalized medicine or precision medicine. Bioinformatics could play a significant role in the development of breakthrough AIT methodologies and in the advancement of allergen bioinformatics. This will certainly contribute to a better knowledge of allergy diseases and will have a beneficial impact on future research in the field.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available within the article.

Acknowledgments

The author is grateful to the Deanship of Scientific Research, King Saud University, Riyadh, Kingdom of Saudi Arabia for funding through the Vice Deanship of Scientific Research Chairs for supporting this research.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Woodfolk, J.A.; Commins, S.P.; Schuyler, A.J.; Erwin, E.A.; Platts-Mills, T.A. Allergens, sources, particles, and molecules: Why do we make IgE responses? Allergol. Int. 2015, 64, 295–303. [Google Scholar] [CrossRef] [Green Version]
  2. Genuneit, J.; Seibold, A.M.; Apfelbacher, C.J.; Konstantinou, G.N.; Koplin, J.J.; La Grutta, S.; Logan, K.; Perkin, M.R.; Flohr, C. The Task Force ‘Overview of Systematic Reviews in Allergy Epidemiology (OSRAE)’ of the EAACI Interest Group on Epidemiology. Overview of systematic reviews in allergy epidemiology. Allergy 2017, 72, 849–856. [Google Scholar] [CrossRef] [PubMed]
  3. Morgenstern, V.; Zutavern, A.; Cyrys, J.; Brockow, I.; Koletzko, S.; Kramer, U.; Behrendt, H.; Herbarth, O.; von Berg, A.; Bauer, C.P. Atopic diseases, allergic sensitization, and exposure to traffic-related air pollution in children. Am. J. Respir. Crit. Care Med. 2008, 177, 1331–1337. [Google Scholar] [CrossRef] [PubMed]
  4. Sharma, N.; Patiyal, S.; Dhall, A.; Pande, A.; Arora, C.; Raghava, G.P. AlgPred 2.0: An improved method for predicting allergenic proteins and mapping of IgE epitopes. Brief. Bioinform. 2021, 22, bbaa294. [Google Scholar] [CrossRef]
  5. Isaacs, K.K.; Goldsmith, M.-R.; Egeghy, P.; Phillips, K.; Brooks, R.; Hong, T.; Wambaugh, J.F. Characterization and prediction of chemical functions and weight fractions in consumer products. Toxicol. Rep. 2016, 3, 723–732. [Google Scholar] [CrossRef] [Green Version]
  6. Kimber, I.; Basketter, D.A.; Gerberick, G.F.; Ryan, C.A.; Dearman, R.J. Chemical allergy: Translating biology into hazard characterization. Toxicol. Sci. 2011, 120 (Suppl. 1), S238–S268. [Google Scholar] [CrossRef]
  7. Basu, S.; Banik, B. Hypersensitivity: An overview. Immunol. Curr. Res. 2018, 2, 1000105. [Google Scholar]
  8. Edwards, A. History of Allergy. In Allergy Frontiers: Diagnosis and Health Economics; Springer: Berlin/Heidelberg, Germany, 2009; pp. 3–19. [Google Scholar]
  9. Wani, N.A.; Mir, M.A.; Qayoom, H.; Mehraj, U.; Nisar, S.; Sheikh, B.A.; Suhail, S. Gell and coomb’s classification of hypersensitivity. In The Fundamentals of Hypersensitivities and Allergies; Nova Science Publishers Inc.: New York, NY, USA, 2020; pp. 35–74. [Google Scholar]
  10. Mir, M.A.; Mehraj, U.; Nisar, S.; Sheikh, B.A.; Suhail, S.; Qayoom, H. Hypersensitivity reaction. In The Fundamentals of Hypersensitivities and Allergies; Nova Science Publishers Inc.: New York, NY, USA, 2020; pp. 1–34. [Google Scholar]
  11. Platts-Mills, T.A. The continuing effect of the discovery of IgE by Kimishige Ishizaka. J. Allergy Clin. Immunol. 2018, 142, 788–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Amarasekera, M. Immunoglobulin E in health and disease. Asia Pac. Allergy 2011, 1, 12–15. [Google Scholar] [CrossRef] [Green Version]
  13. Lei, D.K.; Grammer, L.C. An overview of allergens. Allergy Asthma Proc. 2019, 40, 362–365. [Google Scholar] [CrossRef]
  14. Baldacci, S.; Maio, S.; Cerrai, S.; Sarno, G.; Baïz, N.; Simoni, M.; Annesi-Maesano, I.; Viegi, G.; Study, H. Allergy and asthma: Effects of the exposure to particulate matter and biological allergens. Respir. Med. 2015, 109, 1089–1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Turnbull, J.; Adams, H.; Gorard, D. The diagnosis and management of food allergy and food intolerances. Aliment. Pharmacol. Ther. 2015, 41, 3–25. [Google Scholar] [CrossRef]
  16. Pramod, S.N. Immunological Basis for the Development of Allergic Diseases-Prevalence, Diagnosis and Treatment Strategies. In Cell Interaction-Molecular and Immunological Basis for Disease Management; IntechOpen: London, UK, 2021. [Google Scholar]
  17. Hossny, E.; Ebisawa, M.; El-Gamal, Y.; Arasi, S.; Dahdah, L.; El-Owaidy, R.; Galvan, C.A.; Lee, B.W.; Levin, M.; Martinez, S. Challenges of managing food allergy in the developing world. World Allergy Organ. J. 2019, 12, 100089. [Google Scholar] [CrossRef] [PubMed]
  18. Loh, W.; Tang, M.L. The epidemiology of food allergy in the global context. Int. J. Environ. Res. Public Health 2018, 15, 2043. [Google Scholar] [CrossRef] [Green Version]
  19. Wheatley, L.M.; Togias, A. Allergic Rhinitis. N. Engl. J. Med. 2015, 372, 456–463. [Google Scholar] [CrossRef] [Green Version]
  20. Hamid, Q.; Tulic, M. Immunobiology of asthma. Annu. Rev. Physiol. 2009, 71, 489–507. [Google Scholar] [CrossRef] [PubMed]
  21. Sicherer, S.H.; Sampson, H.A. Food allergy. J. Allergy Clin. Immunol. 2010, 125, S116–S125. [Google Scholar] [CrossRef]
  22. Incorvaia, C.; Frati, F.; Verna, N.; D’Alò, S.; Motolese, A.; Pucci, S. Allergy and the skin. Clin. Exp. Immunol. 2008, 153, 27–29. [Google Scholar] [CrossRef]
  23. Khan, D.A.; Solensky, R. Drug allergy. J. Allergy Clin. Immunol. 2010, 125, S126–S137.e1. [Google Scholar] [CrossRef]
  24. Golden, D.B. Insect allergy. In Middleton’s Allergy Essentials; Elsevier: Amsterdam, The Netherlands, 2017; pp. 377–393. [Google Scholar]
  25. Kemp, S.F.; Lockey, R.F. Anaphylaxis: A review of causes and mechanisms. J. Allergy Clin. Immunol. 2002, 110, 341–348. [Google Scholar] [CrossRef]
  26. Pawankar, R.; Canonica, G.W.; Holgate, S.T.; Lockey, R.F. Allergic diseases and asthma: A major global health concern. Curr. Opin. Allergy Clin. Immunol. 2012, 12, 39–41. [Google Scholar] [CrossRef]
  27. Wang, Z.; Sundy, J.S.; Foss, C.M.; Barnhart, H.X.; Palmer, S.M.; Allgood, S.D.; Trudeau, E.; Alexander, K.M.; Levesque, M.C. Racial differences in the association of CD14 polymorphisms with serum total IgE levels and allergen skin test reactivity. J. Asthma Allergy 2013, 6, 81–92. [Google Scholar] [CrossRef] [Green Version]
  28. Arbes Jr, S.J.; Gergen, P.J.; Elliott, L.; Zeldin, D.C. Prevalences of positive skin test responses to 10 common allergens in the US population: Results from the third National Health and Nutrition Examination Survey. J. Allergy Clin. Immunol. 2005, 116, 377–383. [Google Scholar] [CrossRef]
  29. Janumpally, S.R.; Feldman, S.R.; Gupta, A.K.; Fleischer, A.B. In the United States, blacks and Asian/Pacific Islanders are more likely than whites to seek medical care for atopic dermatitis. Arch. Dermatol. 2002, 138, 634–637. [Google Scholar] [CrossRef] [Green Version]
  30. Mahdavinia, M.; Tobin, M.C.; Fierstein, J.L.; Andy-Nweye, A.B.; Bilaver, L.A.; Fox, S.; Pappalardo, A.A.; Jiang, J.; Catlin, P.A.; Chura, A. African American Children Are More Likely to Be Allergic to Shellfish and Finfish: Findings from FORWARD, a Multisite Cohort Study. J. Allergy Clin. Immunol. Pract. 2021, 9, 2867–2873.e1. [Google Scholar] [CrossRef] [PubMed]
  31. Holt, P.; Macaubas, C.; Stumbles, P.; Sly, P. The role of allergy in the development of asthma. Nature 1999, 402, 12–17. [Google Scholar] [CrossRef] [PubMed]
  32. Ownby, D.R. Environmental factors versus genetic determinants of childhood inhalant allergies. J. Allergy Clin. Immunol. 1990, 86, 279–287. [Google Scholar] [CrossRef]
  33. Knudsen, T.M.; Rezwan, F.I.; Jiang, Y.; Karmaus, W.; Svanes, C.; Holloway, J.W. Transgenerational and intergenerational epigenetic inheritance in allergic diseases. J. Allergy Clin. Immunol. 2018, 142, 765–772. [Google Scholar]
  34. Dor-Wojnarowska, A.; Liebhart, J.; Miecielica, J.; Rabski, M.; Fal, A.; Samoliński, B.; Nittner-Marszalska, M. The impact of sex and age on the prevalence of clinically relevant sensitization and asymptomatic sensitization in the general population. Arch. Immunol. Ther. Exp. 2017, 65, 253–261. [Google Scholar] [CrossRef] [Green Version]
  35. Fuseini, H.; Newcomb, D.C. Mechanisms driving gender differences in asthma. Curr. Allergy Asthma Rep. 2017, 17, 19. [Google Scholar]
  36. Dharma, C.; Lefebvre, D.; Tran, M.; Lou, W.; Subbarao, P.; Becker, A.; Mandhane, P.; Turvey, S.; Sears, M.; investigators, C.S. Patterns of allergic sensitization and atopic dermatitis from 1 to 3 years: Effects on allergic diseases. Clin. Exp. Allergy 2018, 48, 48–59. [Google Scholar] [CrossRef] [PubMed]
  37. van Aalderen, W.M. Childhood asthma: Diagnosis and treatment. Scientifica 2012, 2012, 674204. [Google Scholar] [CrossRef] [Green Version]
  38. Hammer-Helmich, L.; Linneberg, A.; Thomsen, S.F.; Glümer, C. Association between parental socioeconomic position and prevalence of asthma, atopic eczema and hay fever in children. Scand. J. Public Health 2014, 42, 120–127. [Google Scholar] [CrossRef]
  39. Myers, J.M.B.; Hershey, G.K.K. Eczema in early life: Genetics, the skin barrier, and lessons learned from birth cohort studies. J. Pediatr. 2010, 157, 704–714. [Google Scholar] [CrossRef] [Green Version]
  40. Sicherer, S.H. Clinical aspects of gastrointestinal food allergy in childhood. Pediatrics 2003, 111 (Suppl. 3), 1609–1616. [Google Scholar] [PubMed]
  41. Kim, Y.S.; Kim, H.Y.; Ahn, H.-S.; Sohn, T.S.; Song, J.Y.; Lee, Y.B.; Lee, D.-H.; Lee, J.-I.; Jeong, S.C.; Chae, H.S. The association between tobacco smoke and serum immunoglobulin E levels in Korean adults. Intern. Med. 2017, 56, 2571–2577. [Google Scholar]
  42. Metsälä, J.; Lundqvist, A.; Kaila, M.; Gissler, M.; Klaukka, T.; Virtanen, S.M. Maternal and perinatal characteristics and the risk of cow’s milk allergy in infants up to 2 years of age: A case-control study nested in the Finnish population. Am. J. Epidemiol. 2010, 171, 1310–1316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Bendtsen, P.; Grønbaek, M.; Kjaer, S.; Munk, C.; Linneberg, A.; Tolstrup, J. Alcohol consumption and the risk of self-reported perennial and seasonal allergic rhinitis in young adult women in a population-based cohort study. Clin. Exp. Allergy 2008, 38, 1179–1185. [Google Scholar] [CrossRef]
  44. Cakir, E.; Ersu, R.; Uyan, Z.S.; Oktem, S.; Varol, N.; Karakoc, F.; Karadag, B.; Akyol, M.; Dagli, E. The prevalence and risk factors of asthma and allergic diseases among working adolescents. Asian Pac. J. Allergy Immunol. 2010, 28, 122. [Google Scholar]
  45. Lee, C.; Chuang, H.; Hong, C.; Huang, S.; Chang, Y.; Ko, Y.; Yu, H. Lifetime exposure to cigarette smoking and the development of adult-onset atopic dermatitis. Br. J. Dermatol. 2011, 164, 483–489. [Google Scholar] [CrossRef] [Green Version]
  46. McKeever, T.; Lewis, S.; Smith, C.; Collins, J.; Heatlie, H.; Frischer, M.; Hubbard, R. Siblings, multiple births, and the incidence of allergic disease: A birth cohort study using the West Midlands general practice research database. Thorax 2001, 56, 758–762. [Google Scholar] [CrossRef] [Green Version]
  47. Wang, I.-J.; Guo, Y.L.; Lin, T.-J.; Chen, P.-C.; Wu, Y.-N. GSTM1, GSTP1, prenatal smoke exposure, and atopic dermatitis. Ann. Allergy Asthma Immunol. 2010, 105, 124–129. [Google Scholar] [CrossRef]
  48. Saxon, A.; Diaz-Sanchez, D. Air pollution and allergy: You are what you breathe. Nat. Immunol. 2005, 6, 223–226. [Google Scholar] [CrossRef] [PubMed]
  49. Strand, V.; Svartengren, M.; Rak, S.; Barck, C.; Bylin, G. Repeated exposure to an ambient level of NO2 enhances asthmatic response to a nonsymptomatic allergen dose. Eur. Respir. J. 1998, 12, 6–12. [Google Scholar] [CrossRef] [Green Version]
  50. Just, J.; Segala, C.; Sahraoui, F.; Priol, G.; Grimfeld, A.; Neukirch, F. Short-term health effects of particulate and photochemical air pollution in asthmatic children. Eur. Respir. J. 2002, 20, 899–906. [Google Scholar] [CrossRef] [Green Version]
  51. Lin, S.; Liu, X.; Le, L.H.; Hwang, S.-A. Chronic exposure to ambient ozone and asthma hospital admissions among children. Environ. Health Perspect. 2008, 116, 1725–1730. [Google Scholar] [CrossRef] [PubMed]
  52. Gent, J.F.; Triche, E.W.; Holford, T.R.; Belanger, K.; Bracken, M.B.; Beckett, W.S.; Leaderer, B.P. Association of low-level ozone and fine particles with respiratory symptoms in children with asthma. JAMA 2003, 290, 1859–1867. [Google Scholar] [CrossRef] [Green Version]
  53. Penttinen, P.; Vallius, M.; Tiittanen, P.; Ruuskanen, J.; Pekkanen, J. Source-specific fine particles in urban air and respiratory function among adult asthmatics. Inhal. Toxicol. 2006, 18, 191–198. [Google Scholar] [CrossRef] [PubMed]
  54. Gauderman, W.J.; Avol, E.; Gilliland, F.; Vora, H.; Thomas, D.; Berhane, K.; McConnell, R.; Kuenzli, N.; Lurmann, F.; Rappaport, E. The effect of air pollution on lung development from 10 to 18 years of age. N. Engl. J. Med. 2004, 351, 1057–1067. [Google Scholar] [CrossRef] [Green Version]
  55. Ackermann-Liebrich, U.; Leuenberger, P.; Schwartz, J.; Schindler, C.; Monn, C.; Bolognini, G.; Bongard, J.; Brändli, O.; Domenighetti, G.; Elsasser, S. Lung function and long term exposure to air pollutants in Switzerland. Study on Air Pollution and Lung Diseases in Adults (SAPALDIA) Team. Am. J. Respir. Crit. Care Med. 1997, 155, 122–129. [Google Scholar] [CrossRef]
  56. Brauer, M.; Hoek, G.; Smit, H.; De Jongste, J.; Gerritsen, J.; Postma, D.S.; Kerkhof, M.; Brunekreef, B. Air pollution and development of asthma, allergy and infections in a birth cohort. Eur. Respir. J. 2007, 29, 879–888. [Google Scholar] [CrossRef] [PubMed]
  57. Melén, E.; Nyberg, F.; Lindgren, C.M.; Berglind, N.; Zucchelli, M.; Nordling, E.; Hallberg, J.; Svartengren, M.; Morgenstern, R.; Kere, J. Interactions between glutathione S-transferase P1, tumor necrosis factor, and traffic-related air pollution for development of childhood allergic disease. Environ. Health Perspect. 2008, 116, 1077–1084. [Google Scholar] [CrossRef] [Green Version]
  58. Nagel, G.; Weinmayr, G.; Kleiner, A.; Garcia-Marcos, L.; Strachan, D.P.; Group, I.P.T.S. Effect of diet on asthma and allergic sensitisation in the International Study on Allergies and Asthma in Childhood (ISAAC) Phase Two. Thorax 2010, 65, 516–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Rosenheck, R. Fast food consumption and increased caloric intake: A systematic review of a trajectory towards weight gain and obesity risk. Obes. Rev. 2008, 9, 535–547. [Google Scholar] [CrossRef]
  60. Mai, X.M.; Becker, A.; Liem, J.; Kozyrskyj, A. Fast food consumption counters the protective effect of breastfeeding on asthma in children? Clin. Exp. Allergy 2009, 39, 556–561. [Google Scholar] [CrossRef]
  61. Mi, Y.; Norbäck, D.; Tao, J.; Mi, Y.; Ferm, M. Current asthma and respiratory symptoms among pupils in Shanghai, China: Influence of building ventilation, nitrogen dioxide, ozone, and formaldehyde in classrooms. Indoor Air 2006, 16, 454–464. [Google Scholar] [CrossRef] [PubMed]
  62. Garcia-Marcos, L.; Canflanca, I.M.; Garrido, J.B.; Varela, A.L.-S.; Garcia-Hernandez, G.; Grima, F.G.; Gonzalez-Diaz, C.; Carvajal-Uruena, I.; Arnedo-Pena, A.; Busquets-Monge, R.M. Relationship of asthma and rhinoconjunctivitis with obesity, exercise and Mediterranean diet in Spanish schoolchildren. Thorax 2007, 62, 503–508. [Google Scholar] [CrossRef] [Green Version]
  63. Huang, S.L.; Pan, W.H. Dietary fats and asthma in teenagers: Analyses of the first Nutrition and Health Survey in Taiwan (NAHSIT). Clin. Exp. Allergy 2001, 31, 1875–1880. [Google Scholar] [CrossRef]
  64. Lawson, J.A.; Rennie, D.C.; Dosman, J.A.; Cammer, A.L.; Senthilselvan, A. Obesity, diet, and activity in relation to asthma and wheeze among rural dwelling children and adolescents. J. Obes. 2013, 2013. [Google Scholar] [CrossRef]
  65. Wang, C.S.; Wang, J.; Zhang, X.; Zhang, L.; Zhang, H.P.; Wang, L.; Wood, L.G.; Wang, G. Is the consumption of fast foods associated with asthma or other allergic diseases? Respirology 2018, 23, 901–913. [Google Scholar] [CrossRef] [PubMed]
  66. Ellwood, P.; Asher, M.; Björkstén, B.; Burr, M.; Pearce, N.; Robertson, C. Diet and asthma, allergic rhinoconjunctivitis and atopic eczema symptom prevalence: An ecological analysis of the International Study of Asthma and Allergies in Childhood (ISAAC) data. Eur. Respir. J. 2001, 17, 436–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Lee, S.-C.; Yang, Y.-H.; Chuang, S.-Y.; Liu, S.-C.; Yang, H.-C.; Pan, W.-H. Risk of asthma associated with energy-dense but nutrient-poor dietary pattern in Taiwanese children. Asia Pac. J. Clin. Nutr. 2012, 21, 73–81. [Google Scholar] [PubMed]
  68. Gutiérrez-Delgado, R.I.; Barraza-Villarreal, A.; Escamilla-Núñez, M.C.; Solano-González, M.; Moreno-Macías, H.; Romieu, I. Food consumption and asthma in school children in Cuernavaca, Morelos, Mexico. Salud Publica Mex. 2009, 51, 202–211. [Google Scholar] [CrossRef] [Green Version]
  69. Farchi, S.; Forastiere, F.; Agabiti, N.; Corbo, G.; Pistelli, R.; Fortes, C.; Dell’Orco, V.; Perucci, C. Dietary factors associated with wheezing and allergic rhinitis in children. Eur. Respir. J. 2003, 22, 772–780. [Google Scholar] [CrossRef] [Green Version]
  70. Wickens, K.; Barry, D.; Friezema, A.; Rhodius, R.; Bone, N.; Purdie, G.; Crane, J. Fast foods–are they a risk factor for asthma? Allergy 2005, 60, 1537–1541. [Google Scholar] [CrossRef] [PubMed]
  71. Takaoka, M.; Norback, D. Diet among Japanese female university students and asthmatic symptoms, infections, pollen and furry pet allergy. Respir. Med. 2008, 102, 1045–1054. [Google Scholar] [CrossRef] [Green Version]
  72. Bueso, A.K.; Berntsen, S.; Mowinckel, P.; Andersen, L.F.; Lødrup Carlsen, K.C.; Carlsen, K.H. Dietary intake in adolescents with asthma—Potential for improvement. Pediatr. Allergy Immunol. 2011, 22 Pt. I, 19–24. [Google Scholar] [CrossRef]
  73. Sly, P.D.; Boner, A.L.; Björksten, B.; Bush, A.; Custovic, A.; Eigenmann, P.A.; Gern, J.E.; Gerritsen, J.; Hamelmann, E.; Helms, P.J. Early identification of atopy in the prediction of persistent asthma in children. Lancet 2008, 372, 1100–1106. [Google Scholar] [CrossRef] [Green Version]
  74. Jackson, D.J.; Evans, M.D.; Gangnon, R.E.; Tisler, C.J.; Pappas, T.E.; Lee, W.-M.; Gern, J.E.; Lemanske, R.F., Jr. Evidence for a causal relationship between allergic sensitization and rhinovirus wheezing in early life. Am. J. Respir. Crit. Care Med. 2012, 185, 281–285. [Google Scholar] [CrossRef] [Green Version]
  75. Holt, P.G.; Strickland, D.H.; Sly, P.D. Virus infection and allergy in the development of asthma: What is the connection? Curr. Opin. Allergy Clin. Immunol. 2012, 12, 151–157. [Google Scholar] [CrossRef]
  76. Jackson, D.J.; Gangnon, R.E.; Evans, M.D.; Roberg, K.A.; Anderson, E.L.; Pappas, T.E.; Printz, M.C.; Lee, W.-M.; Shult, P.A.; Reisdorf, E. Wheezing rhinovirus illnesses in early life predict asthma development in high-risk children. Am. J. Respir. Crit. Care Med. 2008, 178, 667–672. [Google Scholar] [CrossRef]
  77. Lemanske, R.F., Jr.; Jackson, D.J.; Gangnon, R.E.; Evans, M.D.; Li, Z.; Shult, P.A.; Kirk, C.J.; Reisdorf, E.; Roberg, K.A.; Anderson, E.L. Rhinovirus illnesses during infancy predict subsequent childhood wheezing. J. Allergy Clin. Immunol. 2005, 116, 571–577. [Google Scholar] [CrossRef]
  78. Jackson, D.J. Early Life Viral Infections and the Development of Asthma–A Target for Asthma Prevention? Curr. Opin. Allergy Clin. Immunol. 2014, 14, 131–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Bisgaard, H.; Hermansen, M.N.; Bønnelykke, K.; Stokholm, J.; Baty, F.; Skytt, N.L.; Aniscenko, J.; Kebadze, T.; Johnston, S.L. Association of bacteria and viruses with wheezy episodes in young children: Prospective birth cohort study. BMJ 2010, 341, c4978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Kloepfer, K.M.; Lee, W.M.; Pappas, T.E.; Kang, T.J.; Vrtis, R.F.; Evans, M.D.; Gangnon, R.E.; Bochkov, Y.A.; Jackson, D.J.; Lemanske, R.F., Jr. Detection of pathogenic bacteria during rhinovirus infection is associated with increased respiratory symptoms and asthma exacerbations. J. Allergy Clin. Immunol. 2014, 133, 1301–1307.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Mills, K.H. Prior Exposure to Bacteria Attenuates Viral Disease of the Respiratory Tract: A Role for IL-17 and Innate Immune Memory? American Thoracic Society: New York, NY, USA, 2014. [Google Scholar]
  82. Calzada, D.; Baos, S.; Cremades, L.; Cardaba, B. New treatments for allergy: Advances in peptide immunotherapy. Curr. Med. Chem. 2018, 25, 2215–2232. [Google Scholar] [CrossRef] [PubMed]
  83. Soyer, O.; Akdis, M.; Ring, J.; Behrendt, H.; Crameri, R.; Lauener, R.; Akdis, C. Mechanisms of peripheral tolerance to allergens. Allergy 2013, 68, 161–170. [Google Scholar] [CrossRef]
  84. Akdis, C.A.; Akdis, M. Mechanisms of immune tolerance to allergens: Role of IL-10 and Tregs. J. Clin. Investig. 2014, 124, 4678–4680. [Google Scholar] [CrossRef] [Green Version]
  85. Shamji, M.H.; Durham, S.R. Mechanisms of allergen immunotherapy for inhaled allergens and predictive biomarkers. J. Allergy Clin. Immunol. 2017, 140, 1485–1498. [Google Scholar] [CrossRef] [Green Version]
  86. Larché, M.; Akdis, C.A.; Valenta, R. Immunological mechanisms of allergen-specific immunotherapy. Nat. Rev. Immunol. 2006, 6, 761–771. [Google Scholar] [CrossRef]
  87. Calzada, D.; Baos, S.; Cremades-Jimeno, L.; Cárdaba, B. Immunological mechanisms in allergic diseases and allergen tolerance: The role of treg cells. J. Immunol. Res. 2018, 2018, 6012053. [Google Scholar] [CrossRef] [PubMed]
  88. Hand, T.W.; Vujkovic-Cvijin, I.; Ridaura, V.K.; Belkaid, Y. Linking the microbiota, chronic disease, and the immune system. Trends Endocrinol. Metab. 2016, 27, 831–843. [Google Scholar] [CrossRef] [Green Version]
  89. Thaiss, C.A.; Levy, M.; Suez, J.; Elinav, E. The interplay between the innate immune system and the microbiota. Curr. Opin. Immunol. 2014, 26, 41–48. [Google Scholar] [CrossRef]
  90. Petrus, N.C.; Henneman, P.; Venema, A.; Mul, A.; van Sinderen, F.; Haagmans, M.; Mook, O.; Hennekam, R.C.; Sprikkelman, A.B.; Mannens, M. Cow’s milk allergy in Dutch children: An epigenetic pilot survey. Clin. Transl. Allergy 2016, 6, 16. [Google Scholar] [CrossRef] [Green Version]
  91. Kozakova, H.; Schwarzer, M.; Tuckova, L.; Srutkova, D.; Czarnowska, E.; Rosiak, I.; Hudcovic, T.; Schabussova, I.; Hermanova, P.; Zakostelska, Z. Colonization of germ-free mice with a mixture of three lactobacillus strains enhances the integrity of gut mucosa and ameliorates allergic sensitization. Cell. Mol. Immunol. 2016, 13, 251–262. [Google Scholar] [CrossRef]
  92. Hapfelmeier, S.; Lawson, M.A.; Slack, E.; Kirundi, J.K.; Stoel, M.; Heikenwalder, M.; Cahenzli, J.; Velykoredko, Y.; Balmer, M.L.; Endt, K. Reversible microbial colonization of germ-free mice reveals the dynamics of IgA immune responses. Science 2010, 328, 1705–1709. [Google Scholar] [CrossRef] [Green Version]
  93. Khor, B.; Gardet, A.; Xavier, R.J. Genetics and pathogenesis of inflammatory bowel disease. Nature 2011, 474, 307–317. [Google Scholar] [CrossRef] [Green Version]
  94. Huang, Y.J.; Marsland, B.J.; Bunyavanich, S.; O’Mahony, L.; Leung, D.Y.; Muraro, A.; Fleisher, T.A. The microbiome in allergic disease: Current understanding and future opportunities—2017 PRACTALL document of the American Academy of Allergy, Asthma & Immunology and the European Academy of Allergy and Clinical Immunology. J. Allergy Clin. Immunol. 2017, 139, 1099–1110. [Google Scholar] [PubMed] [Green Version]
  95. Canani, R.B.; De Filippis, F.; Nocerino, R.; Paparo, L.; Di Scala, C.; Cosenza, L.; Della Gatta, G.; Calignano, A.; De Caro, C.; Laiola, M. Gut microbiota composition and butyrate production in children affected by non-IgE-mediated cow’s milk allergy. Sci. Rep. 2018, 8, 12500. [Google Scholar] [CrossRef] [Green Version]
  96. Wang, M.; Karlsson, C.; Olsson, C.; Adlerberth, I.; Wold, A.E.; Strachan, D.P.; Martricardi, P.M.; Åberg, N.; Perkin, M.R.; Tripodi, S. Reduced diversity in the early fecal microbiota of infants with atopic eczema. J. Allergy Clin. Immunol. 2008, 121, 129–134. [Google Scholar] [CrossRef] [PubMed]
  97. Abrahamsson, T.R.; Jakobsson, H.E.; Andersson, A.F.; Björkstén, B.; Engstrand, L.; Jenmalm, M.C. Low diversity of the gut microbiota in infants with atopic eczema. J. Allergy Clin. Immunol. 2012, 129, 434–440.e2. [Google Scholar] [CrossRef] [Green Version]
  98. Huang, Y.J.; Boushey, H.A. The microbiome in asthma. J. Allergy Clin. Immunol. 2015, 135, 25–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Byrd, A.L.; Belkaid, Y.; Segre, J.A. The human skin microbiome. Nat. Rev. Microbiol. 2018, 16, 143–155. [Google Scholar] [CrossRef] [PubMed]
  100. Paller, A.S.; Kong, H.H.; Seed, P.; Naik, S.; Scharschmidt, T.C.; Gallo, R.L.; Luger, T.; Irvine, A.D. The microbiome in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2019, 143, 26–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Nowak-Wegrzyn, A.; Szajewska, H.; Lack, G. Food allergy and the gut. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 241–257. [Google Scholar] [CrossRef]
  102. Sampson, H.A.; O’Mahony, L.; Burks, A.W.; Plaut, M.; Lack, G.; Akdis, C.A. Mechanisms of food allergy. J. Allergy Clin. Immunol. 2018, 141, 11–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Arrieta, M.-C.; Stiemsma, L.T.; Dimitriu, P.A.; Thorson, L.; Russell, S.; Yurist-Doutsch, S.; Kuzeljevic, B.; Gold, M.J.; Britton, H.M.; Lefebvre, D.L. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci. Transl. Med. 2015, 7, 307ra152. [Google Scholar] [CrossRef]
  104. Fujimura, K.E.; Sitarik, A.R.; Havstad, S.; Lin, D.L.; Levan, S.; Fadrosh, D.; Panzer, A.R.; LaMere, B.; Rackaityte, E.; Lukacs, N.W. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat. Med. 2016, 22, 1187–1191. [Google Scholar] [CrossRef] [Green Version]
  105. Hilty, M.; Burke, C.; Pedro, H.; Cardenas, P.; Bush, A.; Bossley, C.; Davies, J.; Ervine, A.; Poulter, L.; Pachter, L. Disordered microbial communities in asthmatic airways. PLoS ONE 2010, 5, e8578. [Google Scholar] [CrossRef] [Green Version]
  106. Fazlollahi, M.; Chun, Y.; Grishin, A.; Wood, R.A.; Burks, A.W.; Dawson, P.; Jones, S.M.; Leung, D.Y.; Sampson, H.A.; Sicherer, S.H. Early-life gut microbiome and egg allergy. Allergy 2018, 73, 1515–1524. [Google Scholar] [CrossRef]
  107. Ege, M.; Mayer, M.; Schwaiger, K.; Mattes, J.; Pershagen, G.; Van Hage, M.; Scheynius, A.; Bauer, J.; Von Mutius, E. Environmental bacteria and childhood asthma. Allergy 2012, 67, 1565–1571. [Google Scholar] [CrossRef]
  108. Vickers, N.J. Animal communication: When I’m calling you, will you answer too? Curr. Biol. 2017, 27, R713–R715. [Google Scholar] [CrossRef] [PubMed]
  109. Abrahamsson, T.; Jakobsson, H.; Andersson, A.F.; Björkstén, B.; Engstrand, L.; Jenmalm, M. Low gut microbiota diversity in early infancy precedes asthma at school age. Clin. Exp. Allergy 2014, 44, 842–850. [Google Scholar] [CrossRef] [Green Version]
  110. Eidelman, A.I.; Schanler, R.J. Breastfeeding and the use of human milk. Pediatrics 2012, 129, e827–e841. [Google Scholar]
  111. Matheson, M.; Allen, K.; Tang, M. Understanding the evidence for and against the role of breastfeeding in allergy prevention. Clin. Exp. Allergy 2012, 42, 827–851. [Google Scholar] [CrossRef] [PubMed]
  112. León-Cava, N.; Lutter, C.; Ross, J.; Martin, L. Quantifying the Benefits of Breastfeeding: A Summary of the Evidence; Pan American Health Organization: Washington, DC, USA, 2002. [Google Scholar]
  113. Oddy, W.H. Breastfeeding, childhood asthma, and allergic disease. Ann. Nutr. Metab. 2017, 70 (Suppl. 2), 26–36. [Google Scholar] [CrossRef] [PubMed]
  114. Muraro, A.; Dreborg, S.; Halken, S.; Høst, A.; Niggemann, B.; Aalberse, R.; Arshad, S.H.; Berg, A.v.; Carlsen, K.H.; Duschén, K. Dietary prevention of allergic diseases in infants and small children: Part III: Critical review of published peer-reviewed observational and interventional studies and final recommendations. Pediatric Allergy Immunol. 2004, 15, 291–307. [Google Scholar] [CrossRef]
  115. Gdalevich, M.; Mimouni, D.; Mimouni, M. Breast-feeding and the risk of bronchial asthma in childhood: A systematic review with meta-analysis of prospective studies. J. Pediatr. 2001, 139, 261–266. [Google Scholar] [CrossRef]
  116. Friedman, N.J.; Zeiger, R.S. The role of breast-feeding in the development of allergies and asthma. J. Allergy Clin. Immunol. 2005, 115, 1238–1248. [Google Scholar] [CrossRef]
  117. Kramer, M.S.; Matush, L.; Vanilovich, I.; Platt, R.; Bogdanovich, N.; Sevkovskaya, Z.; Dzikovich, I.; Shishko, G.; Mazer, B. Effect of prolonged and exclusive breast feeding on risk of allergy and asthma: Cluster randomised trial. Bmj 2007, 335, 815. [Google Scholar] [CrossRef] [Green Version]
  118. Wright, A.; Holberg, C.; Taussig, L.; Martinez, F. Factors influencing the relation of infant feeding to asthma and recurrent wheeze in childhood. Thorax 2001, 56, 192–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Mihrshahi, S.; Ampon, R.; Webb, K.; Almqvist, C.; Kemp, A.; Hector, D.; Marks, G.B.; Team, C. The association between infant feeding practices and subsequent atopy among children with a family history of asthma. Clin. Exp. Allergy 2007, 37, 671–679. [Google Scholar] [CrossRef] [PubMed]
  120. Sears, M.R.; Greene, J.M.; Willan, A.R.; Taylor, D.R.; Flannery, E.M.; Cowan, J.O.; Herbison, G.P.; Poulton, R. Long-term relation between breastfeeding and development of atopy and asthma in children and young adults: A longitudinal study. Lancet 2002, 360, 901–907. [Google Scholar] [CrossRef]
  121. Milani, C.; Duranti, S.; Bottacini, F.; Casey, E.; Turroni, F.; Mahony, J.; Belzer, C.; Delgado Palacio, S.; Arboleya Montes, S.; Mancabelli, L. The first microbial colonizers of the human gut: Composition, activities, and health implications of the infant gut microbiota. Microbiol. Mol. Biol. Rev. 2017, 81, e00036-17. [Google Scholar] [CrossRef] [Green Version]
  122. Bunyavanich, S.; Berin, M.C. Food allergy and the microbiome: Current understandings and future directions. J. Allergy Clin. Immunol. 2019, 144, 1468–1477. [Google Scholar] [CrossRef] [PubMed]
  123. Atarashi, K.; Tanoue, T.; Shima, T.; Imaoka, A.; Kuwahara, T.; Momose, Y.; Cheng, G.; Yamasaki, S.; Saito, T.; Ohba, Y. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011, 331, 337–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Russell, S.L.; Gold, M.J.; Hartmann, M.; Willing, B.P.; Thorson, L.; Wlodarska, M.; Gill, N.; Blanchet, M.R.; Mohn, W.W.; McNagny, K.M. Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Rep. 2012, 13, 440–447. [Google Scholar] [CrossRef] [PubMed]
  125. Geuking, M.B.; Cahenzli, J.; Lawson, M.A.; Ng, D.C.; Slack, E.; Hapfelmeier, S.; McCoy, K.D.; Macpherson, A.J. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity 2011, 34, 794–806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly-y, M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [Green Version]
  127. Feehley, T.; Plunkett, C.H.; Bao, R.; Hong, S.M.C.; Culleen, E.; Belda-Ferre, P.; Campbell, E.; Aitoro, R.; Nocerino, R.; Paparo, L. Healthy infants harbor intestinal bacteria that protect against food allergy. Nat. Med. 2019, 25, 448–453. [Google Scholar] [CrossRef] [PubMed]
  128. Abdel-Gadir, A.; Stephen-Victor, E.; Gerber, G.K.; Rivas, M.N.; Wang, S.; Harb, H.; Wang, L.; Li, N.; Crestani, E.; Spielman, S. Microbiota therapy acts via a regulatory T cell MyD88/RORγt pathway to suppress food allergy. Nat. Med. 2019, 25, 1164–1174. [Google Scholar] [CrossRef]
  129. Berin, M.C. Dysbiosis in food allergy and implications for microbial therapeutics. J. Clin. Investig. 2021, 131, e144994. [Google Scholar] [CrossRef]
  130. Gern, J.E.; Rosenthal, L.A.; Sorkness, R.L.; Lemanske Jr, R.F. Effects of viral respiratory infections on lung development and childhood asthma. J. Allergy Clin. Immunol. 2005, 115, 668–674. [Google Scholar] [CrossRef] [PubMed]
  131. Wark, P.; Johnston, S.; Moric, I.; Simpson, J.; Hensley, M.; Gibson, P. Neutrophil degranulation and cell lysis is associated with clinical severity in virus-induced asthma. Eur. Respir. J. 2002, 19, 68–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Holtzman, M.J.; Morton, J.D.; Shornick, L.P.; Tyner, J.W.; O’Sullivan, M.P.; Antao, A.; Lo, M.; Castro, M.; Walter, M.J. Immunity, inflammation, and remodeling in the airway epithelial barrier: Epithelial-viral-allergic paradigm. Physiol. Rev. 2002, 82, 19–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Gavala, M.L.; Bashir, H.; Gern, J.E. Virus/allergen interactions in asthma. Curr. Allergy Asthma Rep. 2013, 13, 298–307. [Google Scholar] [CrossRef] [Green Version]
  134. Fahy, J.V. Type 2 inflammation in asthma—Present in most, absent in many. Nat. Rev. Immunol. 2015, 15, 57–65. [Google Scholar] [CrossRef]
  135. Bizzintino, J.; Lee, W.; Laing, I.; Vang, F.; Pappas, T.; Zhang, G.; Martin, A.; Khoo, S.; Cox, D.; Geelhoed, G. Association between human rhinovirus C and severity of acute asthma in children. Eur. Respir. J. 2011, 37, 1037–1042. [Google Scholar] [CrossRef]
  136. Sigurs, N.; Gustafsson, P.M.; Bjarnason, R.; Lundberg, F.; Schmidt, S.; Sigurbergsson, F.; Kjellman, B. Severe respiratory syncytial virus bronchiolitis in infancy and asthma and allergy at age 13. Am. J. Respir. Crit. Care Med. 2005, 171, 137–141. [Google Scholar] [CrossRef]
  137. Khetsuriani, N.; Kazerouni, N.N.; Erdman, D.D.; Lu, X.; Redd, S.C.; Anderson, L.J.; Teague, W.G. Prevalence of viral respiratory tract infections in children with asthma. J. Allergy Clin. Immunol. 2007, 119, 314–321. [Google Scholar] [CrossRef]
  138. Arden, K.E.; Chang, A.B.; Lambert, S.B.; Nissen, M.D.; Sloots, T.P.; Mackay, I.M. Newly identified respiratory viruses in children with asthma exacerbation not requiring admission to hospital. J. Med. Virol. 2010, 82, 1458–1461. [Google Scholar] [CrossRef] [PubMed]
  139. Jackson, D.J.; Makrinioti, H.; Rana, B.M.; Shamji, B.W.; Trujillo-Torralbo, M.-B.; Footitt, J.; Del-Rosario, J.; Telcian, A.G.; Nikonova, A.; Zhu, J. IL-33–dependent type 2 inflammation during rhinovirus-induced asthma exacerbations in vivo. Am. J. Respir. Crit. Care Med. 2014, 190, 1373–1382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Beale, J.; Jayaraman, A.; Jackson, D.J.; Macintyre, J.D.; Edwards, M.R.; Walton, R.P.; Zhu, J.; Ching, Y.M.; Shamji, B.; Edwards, M. Rhinovirus-induced IL-25 in asthma exacerbation drives type 2 immunity and allergic pulmonary inflammation. Sci. Transl. Med. 2014, 6, 256ra134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Edwards, M.R.; Strong, K.; Cameron, A.; Walton, R.P.; Jackson, D.J.; Johnston, S.L. Viral infections in allergy and immunology: How allergic inflammation influences viral infections and illness. J. Allergy Clin. Immunol. 2017, 140, 909–920. [Google Scholar] [CrossRef] [Green Version]
  142. Mehmood, I.; Ijaz, M.; Ahmad, S.; Ahmed, T.; Bari, A.; Abro, A.; Allemailem, K.S.; Almatroudi, A.; Tahir ul Qamar, M. SARS-CoV-2: An update on genomics, risk assessment, potential therapeutics and vaccine development. Int. J. Environ. Res. Public Health 2021, 18, 1626. [Google Scholar] [CrossRef]
  143. Alamri, M.A.; ul Qamar, M.T.; Afzal, O.; Alabbas, A.B.; Riadi, Y.; Alqahtani, S.M. Discovery of anti-MERS-CoV small covalent inhibitors through pharmacophore modeling, covalent docking and molecular dynamics simulation. J. Mol. Liq. 2021, 330, 115699. [Google Scholar] [CrossRef]
  144. Hosoki, K.; Chakraborty, A.; Sur, S. Molecular mechanisms and epidemiology of COVID-19 from an allergist’s perspective. J. Allergy Clin. Immunol. 2020, 146, 285–299. [Google Scholar] [CrossRef]
  145. Chhiba, K.D.; Patel, G.B.; Vu, T.H.T.; Chen, M.M.; Guo, A.; Kudlaty, E.; Mai, Q.; Yeh, C.; Muhammad, L.N.; Harris, K.E. Prevalence and characterization of asthma in hospitalized and nonhospitalized patients with COVID-19. J. Allergy Clin. Immunol. 2020, 146, 307–314.e4. [Google Scholar] [CrossRef]
  146. Recalcati, S. Cutaneous manifestations in COVID-19: A first perspective. J. Eur. Acad. Derm. Venereol 2020, 34, e212–e213. [Google Scholar] [CrossRef]
  147. Malipiero, G.; Heffler, E.; Pelaia, C.; Puggioni, F.; Racca, F.; Ferri, S.; Spinello, L.; Merigo, M.; Lamacchia, D.; Cataldo, G. Allergy clinics in times of the SARS-CoV-2 pandemic: An integrated model. Clin. Transl. Allergy 2020, 10, 23. [Google Scholar] [CrossRef] [PubMed]
  148. Cuffel, B.; Wamboldt, M.; Borish, L.; Kennedy, S.; Crystal-Peters, J. Economic consequences of comorbid depression, anxiety, and allergic rhinitis. Psychosomatics 1999, 40, 491–496. [Google Scholar] [CrossRef]
  149. Goodwin, R.D.; Castro, M.; Kovacs, M. Major depression and allergy: Does neuroticism explain the relationship? Psychosom. Med. 2006, 68, 94–98. [Google Scholar] [CrossRef] [Green Version]
  150. Goodwin, R.D.; Galea, S.; Perzanowski, M.; Jacobi, F. Impact of allergy treatment on the association between allergies and mood and anxiety in a population sample. Clin. Exp. Allergy 2012, 42, 1765–1771. [Google Scholar] [CrossRef]
  151. Gonzalez-Diaz, S.N.; Martin, B.; Villarreal-Gonzalez, R.V.; de Lira-Quezada, C.E.; Macouzet-Sanchez, C.; Macias-Weinmann, A.; Guzman-Avilan, R.I.; Garcia-Campa, M.; Noyola-Perez, A.; Garcia-Gonzalez, D.U. Psychological impact of the COVID-19 pandemic on patients with allergic diseases. World Allergy Organ. J. 2021, 14, 100510. [Google Scholar] [CrossRef] [PubMed]
  152. Camacho-Rivera, M.; Islam, J.Y.; Vidot, D.C. Associations between chronic health conditions and COVID-19 preventive behaviors among a nationally representative sample of US adults: An analysis of the COVID impact survey. Health Equity 2020, 4, 336–344. [Google Scholar] [CrossRef]
  153. Galea, S.; Merchant, R.M.; Lurie, N. The mental health consequences of COVID-19 and physical distancing: The need for prevention and early intervention. JAMA Intern. Med. 2020, 180, 817–818. [Google Scholar] [CrossRef] [Green Version]
  154. Tzeng, N.-S.; Chang, H.-A.; Chung, C.-H.; Kao, Y.-C.; Chang, C.-C.; Yeh, H.-W.; Chiang, W.-S.; Chou, Y.-C.; Chang, S.-Y.; Chien, W.-C. Increased risk of psychiatric disorders in allergic diseases: A nationwide, population-based, cohort study. Front. Psychiatry 2018, 9, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Olaguibel, J.M.; Alobid, I.; Alvarez Puebla, M.; Crespo-Lessmann, A.; Domínguez Ortega, J.; García-Rio, F.; Izquierdo-Domínguez, A.; Mullol, J.; Plaza, V.; Quirce, S. Upper and lower airways functional examination in asthma and respiratory allergic deseases. Considerations in the SARS-CoV-2 post-pandemic situation. J. Investig. Allergol. Clin. Immunol. 2020, 31, 17–35. [Google Scholar] [CrossRef]
  156. Izquierdo-Domínguez, A.; Rojas-Lechuga, M.J.; Alobid, I. Management of Allergic Diseases during COVID-19 Outbreak. Curr. Allergy Asthma Rep. 2021, 21, 8. [Google Scholar] [CrossRef] [PubMed]
  157. Kim, Y.-J.; Kim, H.-J.; Kang, M.-J.; Yu, H.-S.; Seo, J.-H.; Kim, H.-Y.; Park, S.-J.; Lee, Y.-C.; Hong, S.-J. Bacillus Calmette-Guérin suppresses asthmatic responses via CD4+ CD25+ regulatory T cells and dendritic cells. Allergy Asthma Immunol. Res. 2014, 6, 201–207. [Google Scholar] [CrossRef]
  158. Choi, I.S. Immunomodulating approach to asthma using mycobacteria. Allergy Asthma Immunol. Res. 2014, 6, 187–188. [Google Scholar] [CrossRef] [Green Version]
  159. Choi, I.S.; Koh, Y.I. Therapeutic effects of BCG vaccination in adult asthmatic patients: A randomized, controlled trial. Ann. Allergy Asthma Immunol. 2002, 88, 584–591. [Google Scholar] [CrossRef]
  160. Chandler, C.E.; Ernst, R.K. Bacterial lipids: Powerful modifiers of the innate immune response. F1000Research 2017, 6, F1000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Bach, J.-F. The effect of infections on susceptibility to autoimmune and allergic diseases. N. Engl. J. Med. 2002, 347, 911–920. [Google Scholar] [CrossRef]
  162. Ribet, D.; Cossart, P. How bacterial pathogens colonize their hosts and invade deeper tissues. Microbes Infect. 2015, 17, 173–183. [Google Scholar] [CrossRef]
  163. Edwards, M.R.; Bartlett, N.W.; Hussell, T.; Openshaw, P.; Johnston, S.L. The microbiology of asthma. Nat. Rev. Microbiol. 2012, 10, 459–471. [Google Scholar] [CrossRef]
  164. Medina, J.L.; Coalson, J.J.; Brooks, E.G.; Winter, V.T.; Chaparro, A.; Principe, M.F.; Kannan, T.R.; Baseman, J.B.; Dube, P.H. Mycoplasma pneumoniae CARDS toxin induces pulmonary eosinophilic and lymphocytic inflammation. Am. J. Respir. Cell Mol. Biol. 2012, 46, 815–822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Chua, K.J.; Kwok, W.C.; Aggarwal, N.; Sun, T.; Chang, M.W. Designer probiotics for the prevention and treatment of human diseases. Curr. Opin. Chem. Biol. 2017, 40, 8–16. [Google Scholar] [CrossRef]
  166. Chung, K.F. Airway microbial dysbiosis in asthmatic patients: A target for prevention and treatment? J. Allergy Clin. Immunol. 2017, 139, 1071–1081. [Google Scholar] [CrossRef] [Green Version]
  167. West, C.E.; Dzidic, M.; Prescott, S.L.; Jenmalm, M.C. Bugging allergy; role of pre-, pro-and synbiotics in allergy prevention. Allergol. Int. 2017, 66, 529–538. [Google Scholar] [CrossRef] [PubMed]
  168. Seggev, J.S.; Sedmak, G.V.; Kurup, V.P. Isotype-specific antibody responses to acute Mycoplasma pneumoniae infection. Ann. Allergy Asthma Immunol. 1996, 77, 67–73. [Google Scholar] [CrossRef]
  169. Darveaux, J.I.; Lemanske, R.F., Jr. Infection-related asthma. J. Allergy Clin. Immunol. Pract. 2014, 2, 658–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Barnes, P. Intrinsic asthma: Not so different from allergic asthma but driven by superantigens? Clin. Exp. Allergy 2009, 39, 1145–1151. [Google Scholar] [CrossRef]
  171. Hahn, D.L.; Schure, A.; Patel, K.; Childs, T.; Drizik, E.; Webley, W. Chlamydia pneumoniae-specific IgE is prevalent in asthma and is associated with disease severity. PLoS ONE 2012, 7, e35945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Patel, K.K.; Anderson, E.; Salva, P.S.; Webley, W.C. The prevalence and identity of Chlamydia-specific IgE in children with asthma and other chronic respiratory symptoms. Respir. Res. 2012, 13, 32. [Google Scholar] [CrossRef] [Green Version]
  173. Hahn, D.L.; Peeling, R.W. Airflow limitation, asthma, and Chlamydia pneumoniae-specific heat shock protein 60. Ann. Allergy Asthma Immunol. 2008, 101, 614–618. [Google Scholar] [CrossRef]
  174. Webley, W.; Tilahun, Y.; Lay, K.; Patel, K.; Stuart, E.; Andrzejewski, C.; Salva, P. Occurrence of Chlamydia trachomatis and Chlamydia pneumoniae in paediatric respiratory infections. Eur. Respir. J. 2009, 33, 360–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Bisgaard, H.; Hermansen, M.N.; Buchvald, F.; Loland, L.; Halkjaer, L.B.; Bønnelykke, K.; Brasholt, M.; Heltberg, A.; Vissing, N.H.; Thorsen, S.V. Childhood asthma after bacterial colonization of the airway in neonates. N. Engl. J. Med. 2007, 357, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
  176. Davis, M.F.; Peng, R.D.; McCormack, M.C.; Matsui, E.C. Staphylococcus aureus colonization is associated with wheeze and asthma among US children and young adults. J. Allergy Clin. Immunol. 2015, 135, 811–813.e5. [Google Scholar] [CrossRef] [Green Version]
  177. Inoshima, I.; Inoshima, N.; Wilke, G.A.; Powers, M.E.; Frank, K.M.; Wang, Y.; Wardenburg, J.B. A Staphylococcus aureus pore-forming toxin subverts the activity of ADAM10 to cause lethal infection in mice. Nat. Med. 2011, 17, 1310–1314. [Google Scholar] [CrossRef] [Green Version]
  178. Ye, Q.; Xu, X.-J.; Shao, W.-X.; Pan, Y.-X.; Chen, X.-J. Mycoplasma pneumoniae infection in children is a risk factor for developing allergic diseases. Sci. World J. 2014, 2014, 986527. [Google Scholar] [CrossRef] [Green Version]
  179. Nakamura, Y.; Oscherwitz, J.; Cease, K.B.; Chan, S.M.; Muñoz-Planillo, R.; Hasegawa, M.; Villaruz, A.E.; Cheung, G.Y.; McGavin, M.J.; Travers, J.B. Staphylococcus δ-toxin induces allergic skin disease by activating mast cells. Nature 2013, 503, 397–401. [Google Scholar] [CrossRef] [Green Version]
  180. Smith-Norowitz, T.A.; Chotikanatis, K.; Erstein, D.P.; Perlman, J.; Norowitz, Y.M.; Joks, R.; Durkin, H.G.; Hammerschlag, M.R.; Kohlhoff, S. Chlamydia pneumoniae enhances the Th2 profile of stimulated peripheral blood mononuclear cells from asthmatic patients. Hum. Immunol. 2016, 77, 382–388. [Google Scholar] [CrossRef] [PubMed]
  181. Yeh, J.-J.; Wang, Y.-C.; Hsu, W.-H.; Kao, C.-H. Incident asthma and Mycoplasma pneumoniae: A nationwide cohort study. J. Allergy Clin. Immunol. 2016, 137, 1017–1023.e6. [Google Scholar] [CrossRef]
  182. Hales, B.J.; Chai, L.Y.; Elliot, C.E.; Pearce, L.J.; Zhang, G.; Heinrich, T.K.; Smith, W.-A.; Kusel, M.M.; Holt, P.G.; Sly, P.D. Antibacterial antibody responses associated with the development of asthma in house dust mite-sensitised and non-sensitised children. Thorax 2012, 67, 321–327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Nordengrün, M.; Michalik, S.; Völker, U.; Bröker, B.M.; Gómez-Gascón, L. The quest for bacterial allergens. Int. J. Med. Microbiol. 2018, 308, 738–750. [Google Scholar] [CrossRef] [PubMed]
  184. Dzoro, S.; Mittermann, I.; Resch-Marat, Y.; Vrtala, S.; Nehr, M.; Hirschl, A.M.; Wikberg, G.; Lundeberg, L.; Johansson, C.; Scheynius, A. House dust mites as potential carriers for IgE sensitization to bacterial antigens. Allergy 2018, 73, 115–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Bonyadi, M.; Hassanzadeh, D.; Seyfizadeh, N.; Borzoueisileh, S. Assessment of allergen-specific IgE by immunoblotting method in atopic dermatitis. Eur. Ann. Allergy Clin. Immunol. 2017, 49, 213–219. [Google Scholar] [CrossRef] [Green Version]
  186. Pullerits, T.; Praks, L.; Ristioja, V.; Lötvall, J. Comparison of a nasal glucocorticoid, antileukotriene, and a combination of antileukotriene and antihistamine in the treatment of seasonal allergic rhinitis. J. Allergy Clin. Immunol. 2002, 109, 949–955. [Google Scholar] [CrossRef]
  187. Chad, Z. Allergies in children. Paediatr. Child. Health 2001, 6, 555–566. [Google Scholar] [CrossRef] [Green Version]
  188. Wert, A.; Posa, D.; Tsilochristou, O.; Schwerk, N. Treatment of allergic children—Where is the progress (for the practicing allergist)? Pediatric Allergy Immunol. 2016, 27, 671–681. [Google Scholar] [CrossRef] [PubMed]
  189. Van Bever, H.P.; Lee, B.W.; Shek, L. The future of research in pediatric allergy: What should the focus be? Pediatric Allergy Immunol. 2012, 23, 5–10. [Google Scholar] [CrossRef]
  190. Douglass, J.A.; O’Hehir, R.E. 1. Diagnosis, treatment and prevention of allergic disease: The basics. Med. J. Aust. 2006, 185, 228–233. [Google Scholar] [CrossRef]
  191. Gardner, L.M.; Thien, F.; Douglass, J.A.; Rolland, J.M.; O’Hehir, R.E. Induction of T ‘regulatory’cells by standardized house dust mite immunotherapy: An increase in CD4+ CD25+ interleukin-10+ T cells expressing peripheral tissue trafficking markers. Clin. Exp. Allergy 2004, 34, 1209–1219. [Google Scholar] [CrossRef]
  192. Wong, A.G.; Lomas, J.M. Allergy testing and immunotherapy. Pediatr. Rev. 2019, 40, 219–228. [Google Scholar] [CrossRef]
  193. Scurlock, A.M. Oral and sublingual immunotherapy for treatment of IgE-mediated food allergy. Clin. Rev. Allergy Immunol. 2018, 55, 139–152. [Google Scholar] [CrossRef]
  194. Astarita, C.; Scala, G.; Sproviero, S.; Franzese, A. Effects of enzyme-potentiated desensitization in the treatment of pollinosis: A double-blind placebo-controlled trial. J. Investig. Allergol. Clin. Immunol. 1996, 6, 248–255. [Google Scholar]
  195. Bochner, B.S.; Rothenberg, M.E.; Boyce, J.A.; Finkelman, F. Advances in mechanisms of allergy and clinical immunology in 2012. J. Allergy Clin. Immunol. 2013, 131, 661–667. [Google Scholar] [CrossRef] [PubMed]
  196. Ismail, S.; Shahid, F.; Khan, A.; Bhatti, S.; Ahmad, S.; Naz, A.; Almatroudi, A.; ul Qamar, M.T. Pan-vaccinomics approach towards a universal vaccine candidate against WHO priority pathogens to address growing global antibiotic resistance. Comput. Biol. Med. 2021, 136, 104705. [Google Scholar] [CrossRef] [PubMed]
  197. Ghosh, D.; Gupta-Bhattacharya, S. Allergen bioinformatics: Recent trends and developments. Sel. Work. Bioinform. 2011, 49, 1–14. [Google Scholar] [CrossRef] [Green Version]
  198. George, A.; Georrge, J.J. Viroinformatics: Databases and tools. Recent Trends Sci. Technol. 2019, 2019, 117–126. [Google Scholar]
  199. George, R.; Jacob, S.; Thomas, S.; Georrge, J.J. Approaches for novel drug target identification. In Proceedings of the International Science Symposium on Recent Trends in Science and Technology, New Delhi, India, 6–10 January 2020; Bharti Publications: New Delhi, India, 2017; pp. 399–421. [Google Scholar]
  200. Zhang, Y.; Huang, Y.; Chen, W.-X.; Xu, Z.-M. Identification of key genes in allergic rhinitis by bioinformatics analysis. J. Int. Med. Res. 2021, 49. [Google Scholar] [CrossRef]
  201. Le Chen, L.S.; Ma, Y.; Zheng, C. Hub Genes Identification in a Murine Model of Allergic Rhinitis Based on Bioinformatics Analysis. Front. Genet. 2020, 11, 970. [Google Scholar] [CrossRef] [PubMed]
  202. Deocaris, C.C.; Rumbaoa, R.G.; Gavarra, A.M.; Alinsug, M.V. A Preliminary analysis of potential allergens in a GMO Rice: A Bioinformatics approach. Open J. Bioinform. Biostat. 2020, 4, 12–16. [Google Scholar]
  203. L’Hocine, L.; Pitre, M.; Achouri, A. Detection and identification of allergens from Canadian mustard varieties of Sinapis alba and Brassica juncea. Biomolecules 2019, 9, 489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Li, C.; Kan, A.; Zeng, X. Bioinformatics Analysis in Different Expression Genes and Potential Pathways of CD4+ Cells in Childhood Allergic Asthma. Res. Sq. 2021, preprint. [Google Scholar] [CrossRef]
  205. Qamar, M.T.U.; Saba Ismail, S.A.; Mirza, M.U.; Abbasi, S.W.; Ashfaq, U.A.; Chen, L.-L. Development of a Novel Multi-Epitope Vaccine Against Crimean-Congo Hemorrhagic Fever Virus: An Integrated Reverse Vaccinology, Vaccine Informatics and Biophysics Approach. Front. Immunol. 2021, 12, 669812. [Google Scholar] [CrossRef] [PubMed]
  206. Aalberse, R.C. Assessment of allergen cross-reactivity. Clin. Mol. Allergy 2007, 5, 2. [Google Scholar] [CrossRef] [Green Version]
  207. Aalberse, R.C.; Stadler, B.M. In silico predictability of allergenicity: From amino acid sequence via 3-D structure to allergenicity. Mol. Nutr. Food Res. 2006, 50, 625–627. [Google Scholar] [CrossRef]
  208. Stadler, M.B.; Stadler, B.M. Allergenicity prediction by protein sequence. FASEB J. 2003, 17, 1141–1143. [Google Scholar] [CrossRef]
  209. Zhang, Z.H.; Koh, J.L.; Zhang, G.L.; Choo, K.H.; Tammi, M.T.; Tong, J.C. AllerTool: A web server for predicting allergenicity and allergic cross-reactivity in proteins. Bioinformatics 2007, 23, 504–506. [Google Scholar] [CrossRef]
  210. Schein, C.H.; Ivanciuc, O.; Braun, W. Structural database of allergenic proteins (SDAP). Food Allergy 2006, 117, 257–283. [Google Scholar] [CrossRef]
  211. Muh, H.C.; Tong, J.C.; Tammi, M.T. AllerHunter: A SVM-pairwise system for assessment of allergenicity and allergic cross-reactivity in proteins. PLoS ONE 2009, 4, e5861. [Google Scholar] [CrossRef] [Green Version]
  212. Vijayakumar, S.; Lakshmi, P. A fuzzy inference system for predicting allergenicity and allergic cross-reactivity in proteins. In Proceedings of the 2013 IEEE International Conference on Bioinformatics and Biomedicine, Shanghai, China, 18–21 December 2013; pp. 49–52. [Google Scholar]
  213. Rehman, A.; Ahmad, S.; Shahid, F.; Albutti, A.; Alwashmi, A.S.; Aljasir, M.A.; Alhumeed, N.; Qasim, M.; Ashfaq, U.A.; Tahir ul Qamar, M. Integrated Core Proteomics, Subtractive Proteomics, and Immunoinformatics Investigation to Unveil a Potential Multi-Epitope Vaccine against Schistosomiasis. Vaccines 2021, 9, 658. [Google Scholar] [CrossRef]
  214. Ahmad, S.; Shahid, F.; Tahir ul Qamar, M.; Abbasi, S.W.; Sajjad, W.; Ismail, S.; Alrumaihi, F.; Allemailem, K.S.; Almatroudi, A.; Ullah Saeed, H.F. Immuno-Informatics Analysis of Pakistan-Based HCV Subtype-3a for Chimeric Polypeptide Vaccine Design. Vaccines 2021, 9, 293. [Google Scholar] [CrossRef] [PubMed]
  215. Solanki, D.; Mandaliya, V.; Georrge, J.J. Allergen Bioinformatics: Repositories and Tools to Predict Allergic Proteins. Recent Trends Sci. Technol. 2020, 2020, 162–172. [Google Scholar]
  216. Chapman, M.D.; Pomés, A.; Breiteneder, H.; Ferreira, F. Nomenclature and structural biology of allergens. J. Allergy Clin. Immunol. 2007, 119, 414–420. [Google Scholar] [CrossRef]
  217. Kadam, K.; Karbhal, R.; Jayaraman, V.; Sawant, S.; Kulkarni-Kale, U. AllerBase: A comprehensive allergen knowledgebase. Database 2017, 2017, bax066. [Google Scholar] [CrossRef]
  218. Radauer, C.; Bublin, M.; Wagner, S.; Mari, A.; Breiteneder, H. Allergens are distributed into few protein families and possess a restricted number of biochemical functions. J. Allergy Clin. Immunol. 2008, 121, 847–852.e7. [Google Scholar] [CrossRef] [PubMed]
  219. Goodman, R.E.; Ebisawa, M.; Ferreira, F.; Sampson, H.A.; van Ree, R.; Vieths, S.; Baumert, J.L.; Bohle, B.; Lalithambika, S.; Wise, J. AllergenOnline: A peer-reviewed, curated allergen database to assess novel food proteins for potential cross-reactivity. Mol. Nutr. Food Res. 2016, 60, 1183–1198. [Google Scholar] [CrossRef] [Green Version]
  220. Nakamura, R.; Teshima, R.; Takagi, K.; Sawada, J.-I. Development of Allergen Database for Food Safety (ADFS): An integrated database to search allergens and predict allergenicity. Kokuritsu Iyakuhin Shokuhin Eisei Kenkyujo Hokoku = Bull. Natl. Inst. Health Sci. 2005, 132, 32–36. [Google Scholar]
  221. Kim, C.-K.; Seol, Y.-J.; Lee, D.-J.; Jeong, I.-S.; Yoon, U.-H.; Lee, J.-Y.; Lee, G.-S.; Park, D.-S. AllergenPro: An integrated database for allergenicity analysis and prediction. Bioinformation 2014, 10, 378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Mari, A.; Scala, E.; Palazzo, P.; Ridolfi, S.; Zennaro, D.; Carabella, G. Bioinformatics applied to allergy: Allergen databases, from collecting sequence information to data integration. The Allergome platform as a model. Cell. Immunol. 2006, 244, 97–100. [Google Scholar] [CrossRef]
  223. ul Qamar, M.T.; Ahmad, S.; Fatima, I.; Ahmad, F.; Shahid, F.; Naz, A.; Abbasi, S.W.; Khan, A.; Mirza, M.U.; Ashfaq, U.A. Designing multi-epitope vaccine against Staphylococcus aureus by employing subtractive proteomics, reverse vaccinology and immuno-informatics approaches. Comput. Biol. Med. 2021, 132, 104389. [Google Scholar] [CrossRef]
  224. Brusic, V.; Petrovsky, N.; Gendel, S.; Millot, M.; Gigonzac, O.; Stelman, S. Computational tools for the study of allergens. Allergy 2003, 58, 1083–1092. [Google Scholar] [CrossRef] [Green Version]
  225. Wang, J.; Zhang, D.; Li, J. PREAL: Prediction of allergenic protein by maximum Relevance Minimum Redundancy (mRMR) feature selection. BMC Syst. Biol. 2013, 7, S9. [Google Scholar] [CrossRef] [Green Version]
  226. Dimitrov, I.; Naneva, L.; Doytchinova, I.; Bangov, I. AllergenFP: Allergenicity prediction by descriptor fingerprints. Bioinformatics 2014, 30, 846–851. [Google Scholar] [CrossRef]
  227. Dimitrov, I.; Bangov, I.; Flower, D.R.; Doytchinova, I. AllerTOP v. 2—A server for in silico prediction of allergens. J. Mol. Modeling 2014, 20, 2278. [Google Scholar] [CrossRef]
  228. Maurer-Stroh, S.; Krutz, N.L.; Kern, P.S.; Gunalan, V.; Nguyen, M.N.; Limviphuvadh, V.; Eisenhaber, F.; Gerberick, G.F. AllerCatPro—prediction of protein allergenicity potential from the protein sequence. Bioinformatics 2019, 35, 3020–3027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Kadam, K.; Sawant, S.; Jayaraman, V.; Kulkarni-Kale, U. Databases and Algorithms in Allergen Informatics. In Bioinformatics—Updated Features and Applications; IntechOpen: London, UK, 2016; p. 53. [Google Scholar]
  230. Yang, X.; Yu, X. An introduction to epitope prediction methods and software. Rev. Med. Virol. 2009, 19, 77–96. [Google Scholar] [CrossRef]
  231. Wang, H.-W.; Pai, T.-W. Machine learning-based methods for prediction of linear B-cell epitopes. In Immunoinformatics; Humana Press: New York, NY, USA, 2014; pp. 217–236. [Google Scholar]
  232. Saha, S.; Raghava, G.P. Prediction methods for B-cell epitopes. In Immunoinformatics; Springer: Berlin/Heidelberg, Germany, 2007; pp. 387–394. [Google Scholar]
  233. Jespersen, M.C.; Peters, B.; Nielsen, M.; Marcatili, P. BepiPred-2.0: Improving sequence-based B-cell epitope prediction using conformational epitopes. Nucleic Acids Res. 2017, 45, W24–W29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Singh, H.; Ansari, H.R.; Raghava, G.P. Improved method for linear B-cell epitope prediction using antigen’s primary sequence. PLoS ONE 2013, 8, e62216. [Google Scholar] [CrossRef] [Green Version]
  235. EL-Manzalawy, Y.; Dobbs, D.; Honavar, V. Predicting linear B-cell epitopes using string kernels. J. Mol. Recognit. Interdiscip. J. 2008, 21, 243–255. [Google Scholar] [CrossRef] [Green Version]
  236. Gao, J.; Faraggi, E.; Zhou, Y.; Ruan, J.; Kurgan, L. BEST: Improved prediction of B-cell epitopes from antigen sequences. PLoS ONE 2012, 7, e40104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Yao, B.; Zhang, L.; Liang, S.; Zhang, C. SVMTriP: A method to predict antigenic epitopes using support vector machine to integrate tri-peptide similarity and propensity. PLoS ONE 2012, 7, e45152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Kringelum, J.V.; Lundegaard, C.; Lund, O.; Nielsen, M. Reliable B cell epitope predictions: Impacts of method development and improved benchmarking. PLoS Comput. Biol. 2012, 8, e1002829. [Google Scholar] [CrossRef]
  239. Giacò, L.; Amicosante, M.; Fraziano, M.; Gherardini, P.F.; Ausiello, G.; Helmer-Citterich, M.; Colizzi, V.; Cabibbo, A. B-Pred, a structure based B-cell epitopes prediction server. Adv. Appl. Bioinform. Chem. AABC 2012, 5, 11. [Google Scholar] [PubMed] [Green Version]
  240. Ponomarenko, J.; Bui, H.-H.; Li, W.; Fusseder, N.; Bourne, P.E.; Sette, A.; Peters, B. ElliPro: A new structure-based tool for the prediction of antibody epitopes. BMC Bioinform. 2008, 9, 514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Ansari, H.R.; Raghava, G.P. Identification of conformational B-cell Epitopes in an antigen from its primary sequence. Immunome Res. 2010, 6, 6. [Google Scholar] [CrossRef] [Green Version]
  242. Krawczyk, K.; Liu, X.; Baker, T.; Shi, J.; Deane, C.M. Improving B-cell epitope prediction and its application to global antibody-antigen docking. Bioinformatics 2014, 30, 2288–2294. [Google Scholar] [CrossRef] [PubMed]
  243. Dimitrov, I.; Garnev, P.; Flower, D.R.; Doytchinova, I. EpiTOP—A proteochemometric tool for MHC class II binding prediction. Bioinformatics 2010, 26, 2066–2068. [Google Scholar] [CrossRef] [Green Version]
  244. Bhasin, M.; Raghava, G.P. Prediction of CTL epitopes using QM, SVM and ANN techniques. Vaccine 2004, 22, 3195–3204. [Google Scholar] [CrossRef] [PubMed]
  245. Oyarzún, P.; Ellis, J.J.; Bodén, M.; Kobe, B. PREDIVAC: CD4+ T-cell epitope prediction for vaccine design that covers 95% of HLA class II DR protein diversity. BMC Bioinform. 2013, 14, 52. [Google Scholar] [CrossRef] [PubMed]
  246. Guan, P.; Hattotuwagama, C.K.; Doytchinova, I.A.; Flower, D.R. MHCPred 2.0. Appl. Bioinform. 2006, 5, 55–61. [Google Scholar] [CrossRef] [PubMed]
  247. Karosiene, E.; Rasmussen, M.; Blicher, T.; Lund, O.; Buus, S.; Nielsen, M. NetMHCIIpan-3. 0, a common pan-specific MHC class II prediction method including all three human MHC class II isotypes, HLA-DR, HLA-DP and HLA-DQ. Immunogenetics 2013, 65, 711–724. [Google Scholar] [CrossRef]
Figure 1. Symptoms and risk factors of allergic diseases.
Figure 1. Symptoms and risk factors of allergic diseases.
Ijerph 18 12105 g001
Figure 2. Allergic reaction mechanisms.
Figure 2. Allergic reaction mechanisms.
Ijerph 18 12105 g002
Figure 3. Overview of viral infection and synergistic or additive effects of allergen exposure causing IgE/TH2 and proTH2 inflammation. IL-25R: IL-25 receptor; GRO-a: melanoma growth stimulating activity-a; IL-13R: IL-13 receptor; IL-33R:IL-33 receptor; ENA-78: epithelial-derived neutrophil-activating protein 78; IL-4R: IL-4 receptor.
Figure 3. Overview of viral infection and synergistic or additive effects of allergen exposure causing IgE/TH2 and proTH2 inflammation. IL-25R: IL-25 receptor; GRO-a: melanoma growth stimulating activity-a; IL-13R: IL-13 receptor; IL-33R:IL-33 receptor; ENA-78: epithelial-derived neutrophil-activating protein 78; IL-4R: IL-4 receptor.
Ijerph 18 12105 g003
Figure 4. Allergy inducing mechanisms of bacteria. * Eo, eosinophil; FCεR, high affinity IgE receptor; B, B cell; Bas, basophil; TSLP, thymic stromal lymphopoietin; MC, mast cell; ILC2, innate lymphoid cell type 2; DC, dendritic cell; Th, T helper cell.
Figure 4. Allergy inducing mechanisms of bacteria. * Eo, eosinophil; FCεR, high affinity IgE receptor; B, B cell; Bas, basophil; TSLP, thymic stromal lymphopoietin; MC, mast cell; ILC2, innate lymphoid cell type 2; DC, dendritic cell; Th, T helper cell.
Ijerph 18 12105 g004
Table 1. Symptoms, causes, and prevalence of different allergy types.
Table 1. Symptoms, causes, and prevalence of different allergy types.
Type of AllergySymptomsPrevalenceAffected OrganCausesReference
Allergic rhinatisisSneezing, itchy, watery, and red eyes, stuffy or runny nose, swelling around the eyes.Affects 10–30% of the population worldwideNoseGenetic and environmental factors[19]
AsthmaWheezing, coughing, shortness of breath, and chest tightnessAffects 3 to 9% of the population worldwideAirways of lungsGenetic and environmental factors[20]
Food allergyItchiness, vomiting, swelling of the tongue, hives, diarrhea, low blood pressure, trouble breathingAffects 8% of the population worldwideSkin, respiratory system, gastrointestinal tractImmune response to food[21]
Skin allergyRash, itching, swelling, redness, cracked skin, flaking or scaling of skin, raised bumpsWorldwide, lifetime prevalence of above 20%SkinLatex, food, drugs, water, sunlight, nickel, chemicals, soap, poison oak or poison ivy[22]
Drug allergyItching, rash, fever, facial swelling, hives, shortness of breath, cardiac symptomsAffects 10% of the population worldwideNose, lungs, throat, ear, lining of the stomach, and skinReactions to medications[23]
Insect allergyItching, pain, and swelling and appearance of redness at the sting/bite or surrounding affected areasMany allergic severe cases have been documented with insect bites worldwide; however, there has been no systemic report.Skin, eyes, throat, tongueInsects bite or sting[24]
AnaphylaxisItchy rash, numbness, throat swelling, lightheadedness, shortness of breathAffects 0.05–2% of the population worldwideSkin, nose, throat, lungs, gastrointestinal tractFoods, insects bites, medications[25]
Table 2. Details of allergen-specific databases.
Table 2. Details of allergen-specific databases.
DatabaseURLMaintained byType of Data ArchivedLast UpdateNumber of Allergens/Haptens/EpitopesReference
IUIS Allergenhttp://www.allergen.org
(Accessed on: 19 August 2021)
World Health Organization (WHO)
and
International Union of Immunological Societies (IUIS)
Allergenicity, structure, sequence (isoforms/isoallergens)Updated continuously853[216]
Structural Database of Allergenic Proteins (SDAP)https://fermi.utmb.edu
(Accessed on: 19 August 2021)
Sealy Centre for
Structural Biology,
University of Texas, USA
IgE epitopes, structure, sequence, structural models20131526[210]
AllerBasewww.bioinfo.net.in/AllerBase/Home.html
(Accessed on: 19 August 2021)
Bioinformatics Centre,
Savitribai Phule PuneUniversity, India
Structure and sequence
(cross-links), IgE epitopes, IgE cross-reactivity, experimental evidences of allergenicity, IgE antibody
Updated continuously2311[217]
AllFamhttp://www.meduniwien.ac.at/allfam
(Accessed on: 19 August 2021)
Department of Pathophysiology and Allergy Research, Medical University of Vienna, AustriaCross-link to Pfam database,
allergen family data
2011936[218]
Allergen Onlinehttp://www.allergenonline.Org
(Accessed on: 20 August 2021)
Food Allergy Research and Resource Program (FARRP) at the University of Nebraska-LincolnAllergenicity, sequence20161956[219]
Allergen Database For Food Safety (ADFS)http://allergen.nihs.go.jp/ADFS/
(Accessed on: 20 August 2021)
National Institute of Health, JapanIgE epitopes, sequence, structure, small molecule allergens20162028[220]
AllergenProhttp://nabic.rda.go.kr/allergen
(Accessed on: 20 August 2021)
The National Agricultural Biotechnology Information Center
(NABIC), Korea
IgE epitopes, sequence20152434[221]
Allergomehttp://www.allergome.org
(Accessed on: 20 August 2021)
Centre for Clinical
and Experimental
Allergology, Italy
Sequence (isoforms/isoallergens), clinical, cross-reactivity, structure, epidemiologicallyUpdated continuously3075[222]
Table 3. Tools/servers for allergen/allergenicity prediction.
Table 3. Tools/servers for allergen/allergenicity prediction.
ServerURLMethodEfficiencyReference
AlgPredhttp://www.imtech.res.in/raghava/algpred/
(Accessed on: 21 August 2021)
SVM and allergen sequence features, epitopes, sequence motifsAccuracy: 85%
Specificity: 88.1%
[4]
AllerHunterhttp://tiger.dbs.nus.edu.sg/AllerHunter/
(Accessed on: 21 August 2021)
SVM and iterative pairwise sequence similarityAccuracy: 95.3%
Specificity: 96.41%
[211]
PREALhttp://gmobl.sjtu.edu.cn/PREAL/index.php
(Accessed on: 21 August 2021)
Physicochemical and biochemical descriptors, sequence features, subcellular locations, SVM, and mRMRAccuracy: 93.42%[225]
AllergenFPhttp://ddg-pharmfac.net/AllergenFP/
(Accessed on: 21 August 2021)
Descriptor-based fingerprints of residues, auto and cross-covarianceAccuracy: 88%[226]
AllerTOPhttp://www.ddg-pharmfac.net/AllerTOP
(Accessed on: 22 August 2021)
Machine learning,
sequence based descriptors,
cross and auto -covariance,
Accuracy: 85.3%
Specificity: 88.1%
[227]
AllerCatProhttps://allercatpro.bii.a-star.edu.sg/
(Accessed on: 22 August 2021)
Sequence similarity, structure similarityAccuracy: 84%
Specificity: 67%
[228]
Table 4. Tools/servers for B cell and T cell epitopes prediction.
Table 4. Tools/servers for B cell and T cell epitopes prediction.
TypeServerURL/WebsiteMethodReference
Linear B cell EpitopeABCPred(http://www.imtech.res.in/raghava/abcpred/)
(Accessed on: 25 August 2021)
ANN[232]
BepiPred(http://www.cbs.dtu.dk/services/BepiPred/)
(Accessed on: 25 August 2021)
HMM[233]
LBtope(http://www.imtech.res.in/raghava/lbtope/)
(Accessed on: 25 August 2021)
SVM[234]
BCPreds(http://crdd.osdd.net/raghava/bcepred/)
(Accessed on: 25 August 2021)
SVM[235]
BEST(http://biomine.ece.ualberta.ca/BEST/)
(Accessed on: 25 August 2021)
SVM[236]
SVMTriP(http://sysbio.unl.edu/SVMTriP/)
(Accessed on: 25 August 2021)
SVM[237]
Conformational/Discontinuous B cell EpitopeDiscoTope 2.0(http://www.cbs.dtu.dk/services/DiscoTope/)
(Accessed on: 27 August 2021)
Structure-based method[238]
B-Pred(http://immuno.bio.uniroma2.it/bpred)
(Accessed on: 27 August 2021)
SVM[239]
ElliPro(http://tools.immuneepitope.org/tools/ElliPro)
(Accessed on: 27 August 2021)
Thornton’s method[240]
CBTOPE(http://crdd.osdd.net/raghava/cbtope/)
(Accessed on: 27 August 2021)
SVM[241]
EpiPred(http://opig.stats.ox.ac.uk/webapps/newsabdab/sabpred/epipred/)
(Accessed on: 27 August 2021)
Structure-based method[242]
T cell EpitopeEpiTOP(http://www.pharmfac.net/EpiTOP/)
(Accessed on: 29 August 2021)
QSAR[243]
CTLPred(http://www.imtech.res.in/raghava/ctlpred/index.html)
(Accessed on: 29 August 2021)
ANN, SVM[244]
PREDIVAC(http://predivac.biosci.uq.edu.au/)
(Accessed on: 29 August 2021)
MM[245]
MHCPred(http://www.ddgpharmfac.net/mhcpred/MHCPred/)
(Accessed on: 29 August 2021)
QSAR[246]
NetMHCIIpan-3.0(http://www.cbs.dtu.dk/services/NetMHCIIpan-3.0/)
(Accessed on: 29 August 2021)
ANN[247]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Aldakheel, F.M. Allergic Diseases: A Comprehensive Review on Risk Factors, Immunological Mechanisms, Link with COVID-19, Potential Treatments, and Role of Allergen Bioinformatics. Int. J. Environ. Res. Public Health 2021, 18, 12105. https://doi.org/10.3390/ijerph182212105

AMA Style

Aldakheel FM. Allergic Diseases: A Comprehensive Review on Risk Factors, Immunological Mechanisms, Link with COVID-19, Potential Treatments, and Role of Allergen Bioinformatics. International Journal of Environmental Research and Public Health. 2021; 18(22):12105. https://doi.org/10.3390/ijerph182212105

Chicago/Turabian Style

Aldakheel, Fahad M. 2021. "Allergic Diseases: A Comprehensive Review on Risk Factors, Immunological Mechanisms, Link with COVID-19, Potential Treatments, and Role of Allergen Bioinformatics" International Journal of Environmental Research and Public Health 18, no. 22: 12105. https://doi.org/10.3390/ijerph182212105

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop