Take a look at the Recent articles

Human microbiota and allergy

Gabriela Gama Freire Alberca

Institute of Biomedical Sciences, Department of Microbiology, University of Sao Paulo, Brazil

Maria Notomi Sato

Laboratory of Dermatology and Immunodeficiencies, University of Sao Paulo Medical School, Brazil

Ricardo Wesley Alberca

Laboratory of Dermatology and Immunodeficiencies, University of Sao Paulo Medical School, Brazil

DOI: 10.15761/GIMCI.1000179

Article
Article Info
Author Info
Figures & Data

Abstract

Human microbiota plays an important role in the development of the immune system and food tolerance. The microbiome is a complex ecosystem, in constant regulation, influenced by the diet and environmental stimulus. Although bacterial communities are present in various body sites [airways, gut, mouth and skin], the gastrointestinal is the largest and most studied host-microbiome interface.

The microbiota is fundamental for maintaining homeostasis at the intestinal barrier and gut dysbiosis can contribute to inflammatory diseases.

Allergy is one of the most common inflammatory disorders, characterized by a hypersensitivity to a harmless substance, and influenced by genetic and environmental factors, including the microbiota.

Here, we aim to review evidences on microbiome´s influence on allergic disorders and microbiota-derived treatments.

Key words

microbiota, microbioma, allergy, treatment, prevention

Introduction and Discussion

Human microbiota and health

The human microbiota is composed of Eeucarya, Bacteria, Archaease and Viruses, these microorganisms play a fundamental role in the metabolism and immunity of humans. Alterations in the microbiome are linked to the development of many diseases such as inflammatory bowel disease, obesity and cancer [1].

The human contact with microorganisms begins in the first minutes of life, with the microbiota colonization in the newborn [2], being influenced by contact with microorganisms present at the moment of birth [3], due to the placenta being considerate microbiome-free [4].

The birth condition, vaginal or caesarean (elective or emergency), presents an implication in the development of the microbiota and extensively studied for the influence in the development of allergy and obesity [5-8]

The host´s microbiota is later influenced by sanitary conditions, food, drugs and environmental factors [9]. Therefore, the individual microbiome presents unique characteristics [10,11] and differs in composition according to the site in the human body, such as mouth, skin, gastrointestinal tract, respiratory tract and genitals. This is due to many factors such as: local pH, temperature and nutrients [12].

 The human gastrointestinal (GI) is the largest interface between the host, environmental factors, antigens and microorganisms in the human body. There are approximately 1013-1014 microorganisms in the GI, with greater genomic content that in the human genome [13].

Microbes and humans have a persistent and symbiotic relationship. Commensal microbes are fundamental for human health, regulating many physiological functions, degradation of substances, production of short-chain fat acids [14] and prevention of allergic sensitization [15]. As a result of this symbiotic process, the host and microorganism are sometimes referred to as “superorganism”.

The microbiome´s environment is in constant regulation, being modulated by external microorganisms and other non-bacterial compounds, for example food in the intestinal microbiota and cosmetical products in the skin. Abrupt change and microbial imbalance (dysbiosis) and/or opportunistic microbes can result in inflammatory stimuli and diseases [16].

The influence of the microbiota in the maturation and development of the immune system is well described [17,18], shaping vaccine´s efficacy, tolerance to food [19] and allergy [20].

Allergy

Allergy is a term first used by Clemens von Pirquet to designate a hypersensitivity to substances [21]. Nowadays, around 1 billion people worldwide are allergic, with an increasing prevalence [22]. The most common allergic manifestations are allergic rhinitis, atopic dermatitis, food allergy and allergic asthma [23].

Allergy is an inflammatory syndrome characterized by a Type 2 immunity with T helper (Th) cells and innate lymphoid cells [ILC] that produces high levels of interleukin [IL]-4, IL-5 and IL-13 denominated as Th2 and ILC2. This process also influences the class switch of B cells into IgE-producing B cells [24,25].

The IgE allergen-specific bind to FC receptors in innate immune cells, mainly basophils and mast cells, which upon a second contact with the allergen releases histamine and lipid mediators promoting the immediate allergic reaction and inflammation. It is crucial to highlight that this process can be lethal in an anaphylactic shock [26,27].

In this second contact with the allergen, Th2 specific cells also migrate to the inflammatory site, promoting inflammation and recruitment of other cells, such as eosinophils [28].

The genetic predisposition to the development of allergy syndromes is well established such as atopy, characterized by the predisposition to high IgE production [29,30]. Dysfunction in genes associated with mucosal barriers, Th2-immunity or immunoregulation can also impact the development of allergy  [31-33].

The environmental contribution to allergy development is well established, for example the exposure to many allergens or pollutants [34]. Recently many studies had connected the microbiome in the development and treatment of allergic diseases [35,36].

Microbiota and allergy

In 1989, the hygiene hypothesis was first proposed as a way to explain the growing incidence of allergies, where the Th1/Th17 immune response in bacterial infection could regulate the Th2 allergic immune response [37]. Later, different hypothesis as the counter-regulatory and “old-friends” were proposed [38,39], in summary the immune stimulation via innate immune receptors would regulate the differentiation of Th1/Th2/Th17 and T regulatory cells (Treg) and regulate not only allergy but other immune diseases [40,41].

This theory’s based on observation and correlation in human´s studies [42-44] were tested in experimental model, where innate immune stimulation with bacterial compounds can in fact curb allergy sensitization [27,45,46],  leading to the possibility of using bacterial compounds to regulate established allergic diseases [47-49].

The allergy-microbiota relation is not only retained to local microbiota and local allergy [example intestinal microbiota and food allergy], but also the relation between host microbiota and the development of allergy, for example in the lung-gut axis [50].

It is established that individuals with a low intestinal microbiota diversity or reduced in specific microorganisms are more susceptible to the development of allergic rhinitis [51] and food allergy [52]. In fact, Stefka, et al. shown that Clostridia-containing microbiota is crucial for the impairment of allergic sensitization [15].

The literature is conflicted about the early or later introduction of food and the development of allergies [53-56], although there are evidence that the introduction to common allergenic foods such peanut, egg or cow’s milk at early age (between 3-6 mounts old) in addition to breastfeeding could help to prevent food-specific allergy [57-60]. And usage of antibiotics can lead susceptibility to allergy [53].

The literature is conflicted about the early or later introduction of food and the development of allergies [54–57], although there is evidence that the introduction to common allergenic foods such peanut, egg or cow’s milk at early age (between 3-6 mounts old) in addition to breastfeeding could help to prevent food-specific allergy [58-61].

These observations are in opposition to the usage of formulas as a substitute for maternal’s milk, which has been associated with the induction of milk´s allergy [62]. This is possible due to maternal´s breastfeeding ability to modulate the offspring's immune system and regulate allergen sensitization [63].

Diet is a major determinant of the GI microbiota [64]. High protein and fat consumption lead to a Bacteroides enterotype and a more carbohydrate-centered diet induces a Prevotella enterotype. Although short-term changes in dietary intake can impact the composition of the microbiota the magnitude is modest and not sufficient to change the enterotype [65]. 

High-fat diets (HFD) are a popular choice for weight loss [66] with similar results that a low-fat diet as long as energy restriction is similar [67]. HFD can result in changes in the microbiota and circulating inflammatory markers [68], and reduction in excretion of short-chain fatty acids [SCFA], which could suggest an increase in the risk of GI disorders [69]. This is greater explored by Kim, et al. which shows that high-fat diets are associated with an increase in allergic rhinitis [70].

Dietary changes can play an important role in the prevention and regulation of allergy [71,72]. This immunomodulation can be obtained by dietary intervention trough anti-inflammatory compounds [73,74] or via modulation of the microbiome [75,76].

High fiber diets can impact the microbiota in the GI protecting the development of food allergy [77] and allergic lung inflammation [78]. The abrogation of the Th2 allergic profile occurs through the production of microbiota-derived SCFAs, especially butyrate and propionate, that induces an anti-inflammatory profile via Treg cells [79]. High fiber consumption can also impact hematopoiesis and abrogate allergic lung inflammation, indicating an intestinal–bone marrow–lung axis that impairs allergic development but can also influence other immunological responses [78].

The oral consumption of butyrate can also directly attenuated lung inflammation and mucus production in OVA-challenged mice [79] and regulate the activation of ILC2 [80], via Gpr109a receptor [81]. Interestingly, SCFA not only promotes a regulation of type 2 immunity, but also induces Th1 and Th17 cell to release IL-10 [82], an anti-inflammatory cytokine [83], that can regulate the development of colitis, inflammatory bowel disease, [84], rheumatoid arthritis, psoriasis, and chronic hepatitis C [85].

The early development of allergies in the offspring can also be curbed by high fiber feeding during pregnancy, not only that but Thorburn, et al. suggest a possible epigenetic modification as a possible mechanism for this inhibition [86]. Microbiota´s metabolites can also promote allergic sensitization, as 12,13-diHOME produced by intestinal microorganisms can impair the activity of T regulatory cells and prejudice food tolerance [87].

Probiotics are another mechanism for allergy treatment, as the consumption of Lactobacillus rhamnosus strain GG and Bifidobacterium lactis Bb12 [88, 89]. This phenomenon can be partially explained by the increase in IgA [90] and increased capacity to secrete IFN- γ [91] or IL-10 [92].

As GI microbiome dysbiosis has been linked to the development of different diseases, fecal microbiota transplantation [FMT] is an emerging therapy for the regulation of GI microbiota [93]. FMT consists of the transfer of microorganisms from the stool of a healthy donor into the gastrointestinal tract of a recipient with a disease related to an unhealthy GI microbiome [94,95].

The FMT can be performed by the infusion in the colon or delivery through the upper GI tract and is currently being proposed for the treatment of ulcerative colitis [96] and inflammatory bowel disease [97,98]. Furthermore, the fecal transplant has been used for the treatment of neuroinflammation in an animal model of Parkinson's disease [99].  Although, the process offers risks related to the transfer of multi-resistant bacteria [100]. 

Microbiota transplant from allergic children or health children was able to transfer allergic sensitization do mice [101], therefore indicating a possible treatment for allergy trough FMT.

Conclusion

The microbiota-host interactions present a crucial role in the regulation of immune responses. Further researches are needed to confirm the safety and efficacy in the treatment of diseases via directly or indirectly manipulation of the microbiota.

Funding

São Paulo State Research Support Foundation (FAPESP) Grant: 19 / 02679-7.

References

  1. Zuo T, Ng SC (2018) The gut microbiota in the pathogenesis and therapeutics of inflammatory bowel disease. Front Microbiol 9.
  2. Fouhy F, Watkins C, Hill CJ, O’Shea CA, Nagle B, et al. (2019) Perinatal factors affect the gut microbiota up to four years after birth. Nat Commun 10: 1517.
  3. Dunn AB, Jordan S, Baker BJ, Carlson NS (2017) The maternal infant microbiome: Considerations for labor and birth. MCN Am J Matern Child Nurs 42: 318-325.
  4. de Goffau MCS, Lager U, Sovio F, Gaccioli E, Cook SJ, et al. (2019) Human placenta has no microbiome but can contain potential pathogens. Nature.
  5. Loo EX, Sim JZ, Loy SL, Goh A, Chan YH, et al. (2017) Associations between caesarean delivery and allergic outcomes: Results from the GUSTO study. Ann Allergy Asthma Immunol 118: 636-638.
  6. Mitselou N, Hallberg J, Stephansson O, Almqvist C, Melén E, et al. (2018) Cesarean delivery, preterm birth, and risk of food allergy: Nationwide Swedish cohort study of more than 1 million children. J Allergy Clin Immunol 142: 1510-1514.
  7. Li H, Ye R, Pei L, Ren A, Zheng X, et al. (2014) Caesarean delivery, caesarean delivery on maternal request and childhood overweight: a Chinese birth cohort study of 181 380 children. Pediatr Obes 9: 10-16.
  8. Ajslev TA, Andersen CS, Gamborg M, Sørensen TI, Jess T (2011) Childhood overweight after establishment of the gut microbiota: the role of delivery mode, pre-pregnancy weight and early administration of antibiotics. Int J Obes 35: 522.
  9. Voreades N, Kozil A, Weir TL (2014) Diet and the development of the human intestinal microbiome. Front Microbiol 5: 494.
  10. Savage DC (1977) Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol 31: 107-133.
  11. Lee SM, Kim N, Park JH, Nam RH, Yoon K, et al. (2018) Comparative analysis of ileal and cecal microbiota in aged rats. J Cancer Prev 23: 70.
  12. Fiocchi C, Souza HS (2012) Microbiota intestinal: sua importância e função. J bras med 100: 30-38.
  13. Ley RE, Peterson DA, Gordon JI (2006) Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell 124: 837-848.
  14. Ma N, Guo P, Zhang J, He T, Kim SW, et al. (2018) Nutrients mediate intestinal bacteria–mucosal immune crosstalk. Front Immunol 9: 5.
  15. Stefka AT, Feehley T, Tripathi P, Qiu J, McCoy K, et al. (2014) Commensal bacteria protect against food allergen sensitization. Proc Natl Acad Sci U S A 111: 13145-13150.
  16. Saltzman ET, Palacios T, Thomsen M, Vitetta L (2018) Intestinal microbiome shifts, dysbiosis, inflammation, and non-alcoholic fatty liver disease. Front Microbiol 9: 61.
  17. Pandiyan P, Bhaskaran N, Schneider E, Jayaraman S, Huehn J (2019) Microbiome dependent regulation of Tregs and Th17 cells in mucosa. Front Immunol 10: 426.
  18. Round JL, Mazmanian SK (2009) The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 9: 313.
  19. da Fonseca DM, Hand TW, Han SJ, Gerner MY, Zaretsky AG, et al. Microbiota-dependent sequelae of acute infection compromise tissue-specific immunity. Cell 163: 354-366.
  20. Chiu CY, Chan YL, Tsai MH, Wang CJ, Chiang MH, et al. (2019) Gut microbial dysbiosis is associated with allergen specific IgE responses in young children with airway allergies. World Allergy Organ J 12: 100021.
  21. Huber B (2006)100 years of allergy: Clemens von Pirquet-his idea of allergy and its immanent concept of disease. Wiener klinische Wochenschrift 118: 573-579.
  22. Akdis CA (Eds) (2013) Global Atlas of Asthma.
  23. Galli SJ, Tsai M, Piliponsky AM (2008) The development of allergic inflammation. Nature 454: 445.
  24. Licona-Limón P, Kim LK, Palm NW, Flavell RA (2013) T H 2, allergy and group 2 innate lymphoid cells. Nat Immunol 14: 536.
  25. Doherty TA, Broide DH (2019) Airway innate lymphoid cells in the induction and regulation of allergy. Allergol Int 68: 9-16.
  26. Alberca Custodio RW, Mirotti L, Gomes E, Nunes FP, S Vieira R, et al. (2019) Dendritic cells expressing MyD88 molecule are necessary and sufficient for CpG-mediated inhibition of IgE production in vivo. Cells 8: 1165.
  27. Nunes FP, Alberca‐Custódio RW, Gomes E, Fonseca DM, Yokoyama NH, et al. (2019) TLR9 agonist adsorbed to alum adjuvant prevents asthma‐like responses induced by Blomia tropicalis mite extract. J Leukoc Biol.
  28. Alberca-Custódio RW, Greiffo FR, MacKenzie B, Oliveira-Junior MC, Andrade-Sousa AS, et al. (2016) Aerobic exercise reduces asthma phenotype by modulation of the leukotriene pathway. Front Immunol 7: 237.
  29. Barnes PJ (2001) Molecular mechanisms of atopy. Mediators Inflamm 10: 285-288.
  30. Pearce N, Pekkanen J, Beasley R (1999) How much asthma is really attributable to atopy?. Thorax 54: 268-272.
  31. Vercelli D (2008) Discovering susceptibility genes for asthma and allergy. Nat Rev Immunol 8: 169.
  32. Ober C, Yao TC (2011) The genetics of asthma and allergic disease: a 21st century perspective. Immunol Rev 242: 10-30.
  33. Akhabir L, Sandford AJ (2011) Genome‐wide association studies for discovery of genes involved in asthma. Respirology 16: 396-406.
  34. Levetin E, Van de Water P (2001) Environmental contributions to allergic disease. Curr Allergy Asthma Rep 1: 506-514.
  35. Huang YJ, Marsland BJ, Bunyavanich S, O'Mahony L, Leung DY, et al. (2017) The microbiome in allergic disease: current understanding and future opportunities. J Allergy Clin Immunol 139: 1099-1110.
  36. Nakatsuji T, Gallo RL (2019) The role of the skin microbiome in atopic dermatitis. Ann Allergy Asthma Immunol 122: 263-269.
  37. Strachan DP (1989) Hay fever, hygiene, and household size. BMJ 299: 1259.
  38. Wills-Karp M, Santeliz J, Karp CL (2001) The germless theory of allergic disease: revisiting the hygiene hypothesis. Nat Rev Immunol 1: 69.
  39. Choi IS (2014) Immunomodulating approach to asthma using mycobacteria. Allergy Asthma Immunol Res 6: 187-188.
  40. Okada H, Kuhn C, Feillet H, Bach JF (2010) The ‘hygiene hypothesis’ for autoimmune and allergic diseases: an update. Clin Exp Immunol 160: 1-9.
  41. Scudellari M (2017) News Feature: Cleaning up the hygiene hypothesis. Proc Natl Acad Sci U S A 114: 1433-1436.
  42. van Strien RT, Engel R, Holst O, Bufe A, Eder W, et al. (2004) Microbial exposure of rural school children, as assessed by levels of N-acetyl-muramic acid in mattress dust, and its association with respiratory health. J Allergy Clin Immunol 113: 860-867.
  43. JM Hopkin, T Shirakawa, T Enomoto, S Shimazu (1997) The inverse association between tuberculin responses and atopic disorder. Clinical Science 92: 7.
  44. Koplin JJ, Dharmage SC, Ponsonby AL, Tang ML, Lowe AJ, et al. (2012) Environmental and demographic risk factors for egg allergy in a population‐based study of infants. Allergy 67: 1415-1422.
  45. Mirotti L, Alberca Custódio RW, Gomes E, Rammauro F, de Araujo EF, et al. (2017) CpG-ODN shapes alum adjuvant activity signaling via MyD88 and IL-10. Front Immunol 8: 47.
  46. Fonseca DM, Wowk PF, Paula MO, Gembre AF, Baruffi MD, et al. (2015) Requirement of MyD88 and F as pathways for the efficacy of allergen‐free immunotherapy. Allergy 70: 275-284.
  47. Tada R, Muto S, Iwata T, Hidaka A, Kiyono H, et al. (2017) Attachment of class B CpG ODN onto DOTAP/DC-chol liposome in nasal vaccine formulations augments antigen-specific immune responses in mice. BMC research notes 10: 68.
  48. Vollmer J, Krieg AM (2009) Immunotherapeutic applications of CpG oligodeoxynucleotide TLR9 agonists. Adv Drug Deliv Rev 61: 195-204.
  49. Frati F, Salvatori C, Incorvaia C, Bellucci A, Di Cara G, et al. (2019) The role of the microbiome in asthma: The Gut–Lung axis. Int J Mol Sci 20: 123.
  50. Bisgaard H, Li N, Bonnelykke K, Chawes BL, Skov T, et al. (2011) Reduced diversity of the intestinal microbiota during infancy is associated with increased risk of allergic disease at school age. J Allergy Clin Immunol 128: 646-652.
  51. Ling Z, Li Z, Liu X, Cheng Y, Luo Y, et al. (2014) Altered fecal microbiota composition associated with food allergy in infants. Appl Environ Microbiol 80: 2546-2554.
  52. Russell SL, Gold MJ, Hartmann M, Willing BP, Thorson L, et al. (2012) Early life antibiotic‐driven changes in microbiota enhance susceptibility to allergic asthma. EMBO reports 13: 440-447.
  53. Schiess SA, Grote V, Scaglioni S, Luque V, Martin F, et al. (2011) Introduction of potentially allergenic foods in the infant’s diet during the first year of life in five European countries. Ann Nutr Metab 58: 109-117.
  54. Grote V, Schiess SA, Closa-Monasterolo R, Escribano J, Giovannini M, et al. (2011) The introduction of solid food and growth in the first 2 y of life in formula-fed children: Analysis of data from a European cohort study. Am J Clin Nutr 94: 1785S-1793S.
  55. Chan ES, Cummings C, Atkinson A, Chad Z, Francoeur M-J, et al. (2014) Dietary exposures and allergy prevention in high-risk infants. Allergy Asthma Clin Immunol 10.
  56. Flokstra‐de Blok BM, Dubois AE, Vlieg‐Boerstra BJ, Oude Elberink JN, Raat H, et al. (2010) Health‐related quality of life of food allergic patients: comparison with the general population and other diseases. Allergy 65: 238-244.
  57. Perkin MR, Logan K, Tseng A, Raji B, Ayis S, et al. (2016) Randomized trial of introduction of allergenic foods in breast-fed infants. N Engl J Med 374: 1733-1743.
  58. Katz Y, Rajuan N, Goldberg MR, Eisenberg E, Heyman E, et al. (2010) Early exposure to cow's milk protein is protective against IgE-mediated cow's milk protein allergy. J Allergy Clin Immunol 126: 77-82.
  59. Koplin JJ, Osborne NJ, Wake M, Martin PE, Gurrin LC, et al. (2010) Can early introduction of egg prevent egg allergy in infants? A population-based study. J Allergy Clin Immunol 126: 807-813.
  60. Du Toit G, Katz Y, Sasieni P, Mesher D, Maleki SJ, et al. (2008) Early consumption of peanuts in infancy is associated with a low prevalence of peanut allergy. J Allergy Clin Immunol 122: 984-991.
  61. Kelly E, DunnGalvin G, Murphy BP, O’B Hourihane J (2019) Formula supplementation remains a risk for cow's milk allergy in breast‐fed infants. Pediatr Allergy Immunol.
  62. Victor JR, Muniz BP, Fusaro AE, de Brito CA, Taniguchi EF, et al. (2010) Maternal immunization with ovalbumin prevents neonatal allergy development and up-regulates inhibitory receptor FcγRIIB expression on B cells. BMC immunology 11: 11.
  63. Robertson RC, Manges AR, Finlay BB, Prendergast AJ (2019) The human microbiome and child growth–first 1000 days and beyond. Trends Microbiol 27: 131-147.
  64. Conlon MA, Bird AR (2015) The impact of diet and lifestyle on gut microbiota and human health. Nutrients 7: 17-44.
  65. Hamdy O, Tasabehji MW, Elseaidy T, Tomah S, Ashrafzadeh S, et al. (2018) Fat versus carbohydrate-based energy-restricted diets for weight loss in patients with type 2 diabetes. Curr Diab Rep 18: 128.
  66. Noakes M, Keogh JB, Foster PR, Clifton PM (2005) Effect of an energy-restricted, high-protein, low-fat diet relative to a conventional high-carbohydrate, low-fat diet on weight loss, body composition, nutritional status, and markers of cardiovascular health in obese women. Am J Clin Nutr 81: 1298-1306.
  67. Wan Y, Wang F, Yuan J, Li J, Jiang D, et al. (2019) Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: a 6-month randomised controlled-feeding trial. Gut 18.
  68. Brinkworth GD, Noakes M, Clifton PM, Bird AR (2009) Comparative effects of very low-carbohydrate, high-fat and high-carbohydrate, low-fat weight-loss diets on bowel habit and faecal short-chain fatty acids and bacterial populations. Br J Nutr 101: 1493-1502.
  69. Kim SY, Sim S, Park B, Kim JH, Choi HG (2016) High-fat and low-carbohydrate diets are associated with allergic rhinitis but not asthma or atopic dermatitis in children. PloS one 11: e0150202.
  70. Bunyavanich S (2019) Food allergy: could the gut microbiota hold the key?. Nat Rev Gastroenterol Hepatol 16: 201.
  71. Canani RB, Paparo L, Nocerino R, Di Scala C, Della Gatta G, et al. (2019) Gut microbiome as target for innovative strategies against food allergy. Front Immunol 10.
  72. Van den Elsen LW, Meulenbroek LA, van Esch BC, Hofman GA, Boon L, et al. (2013) CD 25+ regulatory T cells transfer n‐3 long chain polyunsaturated fatty acids‐induced tolerance in mice allergic to cow's milk protein. Allergy 68: 1562-1570.
  73. Iwamura C, Shinoda K, Yoshimura M, Watanabe Y, Obata A, et al. (2010) Naringenin chalcone suppresses allergic asthma by inhibiting the type-2 function of CD4 T cells. Allergol Int 59: 67-73.
  74. Umu ÖC, Rudi K, Diep DB (2017) Modulation of the gut microbiota by prebiotic fibres and bacteriocins. Microb Ecol Health Dis 28: 1348886.
  75. Aitoro R, Paparo L, Amoroso A, Di Costanzo M, Cosenza L, et al. (2017) Gut microbiota as a target for preventive and therapeutic intervention against food allergy. Nutrients 9: 672.
  76. Schouten B, van Esch BC, Hofman GA, de Kivit S, Boon L, et al. (2012) A potential role for CD25+ regulatory T-cells in the protection against casein allergy by dietary non-digestible carbohydrates. Br J Nutr 107: 96-105.
  77. Trompette A, Gollwitzer ES, Pattaroni C, Lopez-Mejia IC, Riva E, et al. (2018) Dietary fiber confers protection against flu by shaping Ly6c− patrolling monocyte hematopoiesis and CD8+ T cell metabolism. Immunity 48: 992-1005.
  78. Vieira RD, Castoldi A, Basso PJ, Hiyane MI, Câmara NO, et al. (2019) Butyrate attenuates lung inflammation by negatively modulating Th9 cells. Front Immunol 10: 67.
  79. Thio CL, Chi PY, Lai AC, Chang YJ (2018) Regulation of type 2 innate lymphoid cell–dependent airway hyperreactivity by butyrate. J Allergy Clin Immunol 142: 1867-1883.
  80. Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, et al. (2014) Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 40: 128-139.
  81. Sun M, Wu W, Chen L, Yang W, Huang X, et al. (2018) Microbiota-derived short-chain fatty acids promote Th1 cell IL-10 production to maintain intestinal homeostasis. Nat Commun 9: 3555.
  82. Moore KW, de Waal Malefyt R, Coffman RL, O'Garra A (2001) Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol 19: 683-765.
  83. Ip WE, Hoshi N, Shouval DS, Snapper S, Medzhitov R (2017) Anti-inflammatory effect of IL-10 mediated by metabolic reprogramming of macrophages. Science 356: 513-519.
  84. Asadullah K, Sterry W, Volk HD (2003) Interleukin-10 therapy—review of a new approach. ‎Pharmacol Rev 55: 241-269.
  85. Thorburn AN, McKenzie CI, Shen S, Stanley D, Macia L, et al. (2015) Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites. Nat Commun 6: 1.
  86. Levan SR, Stamnes KA, Lin DL, Panzer AR, Fukui E, et al. (2019) Elevated faecal 12, 13-diHOME concentration in neonates at high risk for asthma is produced by gut bacteria and impedes immune tolerance. Nat Microbiol.
  87. Savilahti E (2011) Probiotics in the treatment and prevention of allergies in children. Biosci Microflora 30: 119-128.
  88. Majamaa H, Isolauri E (1997) Probiotics: a novel approach in the management of food allergy. J Allergy Clin Immunol 99: 179-185.
  89. Viljanen M, Kuitunen M, Haahtela T, Juntunen‐Backman K, Korpela R, et al. (2005) Probiotic effects on faecal inflammatory markers and on faecal IgA in food allergic atopic eczema/dermatitis syndrome infants. Pediatr Allergy Immunol 16: 65-71.
  90. Pohjavuori E, Viljanen M, Korpela R, Kuitunen M, Tiittanen M, et al. (2004) Lactobacillus GG effect in increasing IFN-γ production in infants with cow's milk allergy. J Allergy Clin Immunol 114: 131-136.
  91. Marschan E, Kuitunen M, Kukkonen K, Poussa T, Sarnesto A, et al.  (2008) Probiotics in infancy induce protective immune profiles that are characteristic for chronic low‐grade inflammation. Clin Exp Allergy 38: 611-618.
  92. West CE, Renz H, Jenmalm MC, Kozyrskyj AL, Allen KJ, et al. (2015) The gut microbiota and inflammatory noncommunicable diseases: associations and potentials for gut microbiota therapies. J Allergy Clin Immunol 135: 3-13.
  93. Kim KO, Gluck M (2019) Fecal microbiota transplantation: An update on clinical practice. Clin Endosc 52: 137.
  94. Moore T, Rodriguez A, Bakken JS (2013) Fecal microbiota transplantation: a practical update for the infectious disease specialist. Clin Infect Dis 58: 541-545.
  95. Costello SP, Soo W, Bryant RV, Jairath V, Hart AL, et al. (2017) Systematic review with meta‐analysis: faecal microbiota transplantation for the induction of remission for active ulcerative colitis.             Aliment Pharmacol Ther 46: 213-224.
  96. Imdad A, Nicholson MR, Tanner‐Smith EE, Zackular JP, Gomez‐Duarte OG, et al. (2018) Fecal transplantation for treatment of inflammatory bowel disease. Cochrane Database Syst Rev 11.
  97. Sunkara T, Rawla P, Ofosu A, Gaduputi V (2018) Fecal microbiota transplant–a new frontier in inflammatory bowel disease. J Inflamm Res 11: 321.
  98. Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, et al. (2016) Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell 167: 1469-1480.
  99. Giles EM, D’Adamo GL, Forster SC (2019) The future of faecal transplants. Nat Rev Microbiol 18: 1.
  100. Abdel-Gadir A, Stephen-Victor E, Gerber GK, Noval RM, Wang S, et al. (2019) Microbiota therapy acts via a regulatory T cell MyD88/RORγt pathway to suppress food allergy. Nat Med 25: 1458.

Editorial Information

Editor-in-Chief

Article Type

Review Article

Publication history

Received date: October 15, 2019
Accepted date: November 28, 2019
Published date: December 02, 2019

Copyright

©2019 Freire Alberca GG. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Freire Alberca GG (2019) Human microbiota and allergy. Gen Int Med Clin Innov 4(3): DOI: 10.15761/GIMCI.1000179

Corresponding author

Ricardo Wesley Alberca

Laboratory of Dermatology and Immunodeficiencies, University of Sao Paulo Medical School, Sao Paulo-SP Brazil

No Figures.