Skip to main content

Advertisement

Log in

Role of the Microbiome in Food Allergy

  • Food Allergy (T Green, Section Editor)
  • Published:
Current Allergy and Asthma Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Resident microbial communities likely modify risk for allergic disorders, including food allergy. We review epidemiologic studies linking microbial exposures to food allergy risk and discuss the mechanisms by which the microbiome may modulate oral tolerance. We additionally address ongoing translational efforts in human microbiome studies.

Recent Findings

Epidemiologic studies and murine models support that altered microbial exposures and colonization in early life modify food allergy risk. Differential microbiota confer protection or susceptibility to food allergy by modulating the regulatory tone of the mucosal immune system. Recent efforts are focused on the identification of bacterial strains necessary for oral tolerance in human and microbial-based clinical trials.

Summary

Early childhood appears to be critical for the colonization of a diverse microbiota necessary for the induction and maintenance of oral tolerance. Identification and functional evaluation of protective commensal microbes will inform strategies for the prevention and treatment of food allergy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Sicherer SH, Sampson HA. Food allergy: a review and update on epidemiology, pathogenesis, diagnosis, prevention, and management. J Allergy Clin Immunol. 2018;141:41–58. https://doi.org/10.1016/j.jaci.2017.11.003.

    Article  CAS  PubMed  Google Scholar 

  2. Osborne NJ, Koplin JJ, Martin PE, et al. Prevalence of challenge-proven IgE-mediated food allergy using population-based sampling and predetermined challenge criteria in infants. J Allergy Clin Immunol. 2011:127, 668–76.e1-2. https://doi.org/10.1016/j.jaci.2011.01.039.

  3. Huang YJ, Marsland BJ, Bunyavanich S, O'Mahony L, Leung DYM, Muraro A, et al. 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;139:1099–110. https://doi.org/10.1016/j.jaci.2017.02.007.

    Article  PubMed  Google Scholar 

  4. Lynch SV, Pedersen O. The human intestinal microbiome in health and disease. N Engl J Med. 2016;375:2369–79. https://doi.org/10.1056/NEJMra1600266.

    Article  CAS  PubMed  Google Scholar 

  5. • Fujimura KE, Sitarik AR, Havstad S, Lin DL, Levan S, Fadrosh D, et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat Med. 2016;22:1187–91. https://doi.org/10.1038/nm.4176. This is one of the first 16S rRNA-based studies showing that compositionally distinct neonatal human gut microbiota exist and are differentially related to relative risk of childhood atopy. In ex vivo experiments, microbiota associated with high atopy risk led to an increased proportion of IL-4 secreting CD4+ T cells and reduced proportion of Treg cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Arrieta M-C, Stiemsma LT, Dimitriu PA, Thorson L, Russell S, Yurist-Doutsch S, et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci Transl Med. 2015;7:307ra152. https://doi.org/10.1126/scitranslmed.aab2271.

    Article  PubMed  Google Scholar 

  7. Stein MM, Hrusch CL, Gozdz J, Igartua C, Pivniouk V, Murray SE, et al. Innate immunity and asthma risk in Amish and Hutterite farm children. N Engl J Med. 2016;375:411–21. https://doi.org/10.1056/NEJMoa1508749.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Williams MR, Gallo RL. The role of the skin microbiome in atopic dermatitis. Curr Allergy Asthma Rep. 2015;15:65. https://doi.org/10.1007/s11882-015-0567-4.

    Article  PubMed  Google Scholar 

  9. Weinstock GM. Genomic approaches to studying the human microbiota. Nature. 2012;489:250–6. https://doi.org/10.1038/nature11553.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Mutius von E, Radon K. Living on a farm: impact on asthma induction and clinical course. Immunol Allergy Clin North Am. 2008;28:631–47– ix–x. https://doi.org/10.1016/j.iac.200803.010.

    Article  Google Scholar 

  11. Waser M, Michels KB, Bieli C, Flöistrup H, Pershagen G, von Mutius E, et al. Inverse association of farm milk consumption with asthma and allergy in rural and suburban populations across Europe. Clin Exp Allergy. 2007;37:661–70. https://doi.org/10.1111/j.1365-2222.2006.02640.x.

    Article  CAS  PubMed  Google Scholar 

  12. Strachan DP. Hay fever, hygiene, and household size. BMJ. 1989;299:1259–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Aichbhaumik N, Zoratti EM, Strickler R, Wegienka G, Ownby DR, Havstad S, et al. Prenatal exposure to household pets influences fetal immunoglobulin E production. Clin Exp Allergy. 2008;38:1787–94. https://doi.org/10.1111/j.1365-2222.2008.03079.x.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Ownby DR, Johnson CC, Peterson EL. Exposure to dogs and cats in the first year of life and risk of allergic sensitization at 6 to 7 years of age. JAMA. 2002;288:963–72.

    Article  PubMed  Google Scholar 

  15. Fujimura KE, Johnson CC, Ownby DR, et al. Man’s best friend? The effect of pet ownership on house dust microbial communities. J Allergy Clin Immunol. 2010:126:410–2– 412.e1–3. https://doi.org/10.1016/j.jaci.201005.042.

  16. Raciborski F, Tomaszewska A, Komorowski J, Samel-Kowalik P, Białoszewski A, Artur W, et al. The relationship between antibiotic therapy in early childhood and the symptoms of allergy in children aged 6-8 years—the questionnaire study results. Int J Occup Med Environ Health. 2012;25:470–80. https://doi.org/10.2478/S13382-012-0056-0.

    Article  PubMed  Google Scholar 

  17. Stensballe LG, Simonsen J, Jensen SM, Bønnelykke K, Bisgaard H. Use of antibiotics during pregnancy increases the risk of asthma in early childhood. J Pediatr. 2013;162:832–838.e3. https://doi.org/10.1016/j.jpeds.2012.09.049.

    Article  CAS  PubMed  Google Scholar 

  18. Murk W, Risnes KR, Bracken MB. Prenatal or early-life exposure to antibiotics and risk of childhood asthma: a systematic review. Pediatrics. 2011;127:1125–38. https://doi.org/10.1542/peds.2010-2092.

    Article  PubMed  Google Scholar 

  19. Sun W, Svendsen ER, Karmaus WJJ, Kuehr J, Forster J. Early-life antibiotic use is associated with wheezing among children with high atopic risk: a prospective European study. J Asthma. 2015;52:647–52. https://doi.org/10.3109/02770903.2014.999284.

    Article  PubMed  Google Scholar 

  20. Koplin JJ, Dharmage SC, Ponsonby A-L, Tang MLK, Lowe AJ, Gurrin LC, et al. Environmental and demographic risk factors for egg allergy in a population-based study of infants. Allergy. 2012;67:1415–22. https://doi.org/10.1111/all.12015.

    Article  CAS  PubMed  Google Scholar 

  21. Tun HM, Konya T, Takaro TK, et al. Exposure to household furry pets influences the gut microbiota of infant at 3–4 months following various birth scenarios. Microbiome. 2017;5:40. https://doi.org/10.1186/s40168-017-0254-x.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Eggesbø M, Botten G, Stigum H, Nafstad P, Magnus P. Is delivery by cesarean section a risk factor for food allergy? J Allergy Clin Immunol. 2003;112:420–6.

    Article  PubMed  Google Scholar 

  23. Metsälä J, Lundqvist A, Kaila M, et al. 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;171:1310–6. https://doi.org/10.1093/aje/kwq074.

    Article  PubMed  Google Scholar 

  24. Bager P, Wohlfahrt J, Westergaard T. Caesarean delivery and risk of atopy and allergic disease: meta-analyses. Clin Exp Allergy. 2008;38:634–42. https://doi.org/10.1111/j.1365-2222.2008.02939.x.

    Article  CAS  PubMed  Google Scholar 

  25. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci U S A. 2010;107:11971–5. https://doi.org/10.1073/pnas.1002601107.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Biasucci G, Rubini M, Riboni S, Morelli L, Bessi E, Retetangos C. Mode of delivery affects the bacterial community in the newborn gut. Early Hum Dev. 2010;86(Suppl 1):13–5. https://doi.org/10.1016/j.earlhumdev.2010.01.004.

    Article  PubMed  Google Scholar 

  27. Jakobsson HE, Abrahamsson TR, Jenmalm MC, Harris K, Quince C, Jernberg C, et al. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut. 2014;63:559–66. https://doi.org/10.1136/gutjnl-2012-303249.

    Article  CAS  PubMed  Google Scholar 

  28. Sepp E, Julge K, Vasar M, Naaber P, Björksten B, Mikelsaar M. Intestinal microflora of Estonian and Swedish infants. Acta Paediatr. 1997;86:956–61.

    Article  CAS  PubMed  Google Scholar 

  29. Björksten B, Naaber P, Sepp E, Mikelsaar M. The intestinal microflora in allergic Estonian and Swedish 2-year-old children. Clin Exp Allergy. 1999;29:342–6.

    Article  PubMed  Google Scholar 

  30. Cahenzli J, Köller Y, Wyss M, Geuking MB, McCoy KD. Intestinal microbial diversity during early-life colonization shapes long-term IgE levels. Cell Host Microbe. 2013;14:559–70. https://doi.org/10.1016/j.chom.2013.10.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Rodriguez B, Prioult G, Bibiloni R, Nicolis I, Mercenier A, Butel MJ, et al. Germ-free status and altered caecal subdominant microbiota are associated with a high susceptibility to cow's milk allergy in mice. FEMS Microbiol Ecol. 2011;76:133–44. https://doi.org/10.1111/j.1574-6941.2010.01035.x.

    Article  CAS  PubMed  Google Scholar 

  32. Bashir MEH, Louie S, Shi HN, Nagler-Anderson C. Toll-like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy. J Immunol. 2004;172:6978–87.

    Article  CAS  PubMed  Google Scholar 

  33. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol. 2009;9:313–23. https://doi.org/10.1038/nri2515.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Noval Rivas M, Burton OT, Wise P, Zhang YQ, Hobson SA, Garcia Lloret M, et al. A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis. J Allergy Clin Immunol. 2013;131:201–12. https://doi.org/10.1016/j.jaci.2012.10.026.

    Article  CAS  PubMed  Google Scholar 

  35. Berni Canani R, Nocerino R, Terrin G, et al. Effect of Lactobacillus GG on tolerance acquisition in infants with cow’s milk allergy: a randomized trial. J Allergy Clin Immunol 2012:129:580–2– 582.e1–5. doi: https://doi.org/10.1016/j.jaci.201110.004.

  36. •• Berni Canani R, Di Costanzo M, Bedogni G, et al. Extensively hydrolyzed casein formula containing Lactobacillus rhamnosus GG reduces the occurrence of other allergic manifestations in children with cow’s milk allergy: 3-year randomized controlled trial. J Allergy Clin Immunol. 2017;139:1906–1913.e4. https://doi.org/10.1016/j.jaci.2016.10.050. This was a 3-year randomized controlled trial in cow’s milk-allergic children to evaluate the co-administration of Lactobacillus rhamnosus GG (LGG) and extensively hydrolyzed casein formula (EHCF). In contrast to the null effect of Lactobacillus casei supplementation reported in another trial [64], LGG+EHCF hastened the development of oral tolerance to cow’s milk and reduced the incidence of at least another atopic disorder during the follow-up period.

    Article  CAS  PubMed  Google Scholar 

  37. Hol J, van Leer EHG, Elink Schuurman BEE, de Ruiter LF, Samsom JN, Hop W, et al. The acquisition of tolerance toward cow’s milk through probiotic supplementation: a randomized, controlled trial. J Allergy Clin Immunol. 2008;121:1448–54. https://doi.org/10.1016/j.jaci.2008.03.018.

    Article  CAS  PubMed  Google Scholar 

  38. •• Bunyavanich S, Shen N, Grishin A, et al. Early-life gut microbiome composition and milk allergy resolution. J Allergy Clin Immunol. 2016;138:1122–30. https://doi.org/10.1016/j.jaci.2016.03.041. This study examined the association between early-life gut microbiota and resolution of cow’s milk allergy. Enrichment of gut Clostridia and Firmicutes at age 3–6 months was associated with milk allergy resolution by age 8 years, suggesting that early infancy is a window during which gut microbiota may shape food allergy outcomes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500:232–6. https://doi.org/10.1038/nature12331.

    Article  CAS  PubMed  Google Scholar 

  40. •• Stefka AT, Feehley T, Tripathi P, Qiu J, McCoy K, Mazmanian SK, et al. Commensal bacteria protect against food allergen sensitization. Proc Natl Acad Sci. 2014;111:13145–50. https://doi.org/10.1073/pnas.1412008111. By selectively colonizing gnotobiotic mice, the group demonstrated that food allergy-protective capacity is conferred by a Clostridia -containing microbiota. Importantly, the authors showed that Clostridia colonization induced intestinal IL-22 production, which resulted in reduced intestinal barrier permeability to food allergens.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. •• Azad MB, Konya T, Guttman DS, et al. Infant gut microbiota and food sensitization: associations in the first year of life. Clin Exp Allergy. 2015;45:632–43. https://doi.org/10.1111/cea.12487. Low gut microbiota richness, overrepresentation of infant gut Enterobacteriaceae , and underrepresentation of Bacteroidaceae at age 3 months were associated with subsequent food sensitization at age 1 year, suggesting that dysbiosis may precede the development of food allergy.

    Article  CAS  PubMed  Google Scholar 

  42. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–41. https://doi.org/10.1126/science.1198469.

    Article  CAS  PubMed  Google Scholar 

  43. Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A. 2010;107:12204–9. https://doi.org/10.1073/pnas.0909122107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sudo N, Sawamura S, Tanaka K, et al. The requirement of intestinal bacterial flora for the development of an IgE production system fully susceptible to oral tolerance induction. J Immunol. 1997;159:1739–45.

    CAS  PubMed  Google Scholar 

  45. • Fazlollahi M, Chun Y, Grishin A, et al. Early-life gut microbiome and egg allergy. Allergy. 2018; https://doi.org/10.1111/all.13389. This study showed a distinct early-life gut microbiome in egg-allergic and egg-sensitized children vs. controls. Genera from the Lachnospiraceae , Streptococcaceae , and Leuconostocaceae families were differentially abundant in children with egg allergy.

  46. Mileti E, Matteoli G, Iliev ID, Rescigno M. Comparison of the immunomodulatory properties of three probiotic strains of Lactobacilli using complex culture systems: prediction for in vivo efficacy. PLoS One. 2009;4:e7056. https://doi.org/10.1371/journal.pone.0007056.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Rodriguez B, Prioult G, Hacini-Rachinel F, Moine D, Bruttin A, Ngom-Bru C, et al. Infant gut microbiota is protective against cow’s milk allergy in mice despite immature ileal T-cell response. FEMS Microbiol Ecol. 2012;79:192–202. https://doi.org/10.1111/j.1574-6941.2011.01207.x.

    Article  CAS  PubMed  Google Scholar 

  48. McCoy KD, Harris NL, Diener P, et al. Natural IgE production in the absence of MHC Class II cognate help. Immunity. 2006;24:329–39. https://doi.org/10.1016/j.immuni.2006.01.013.

    Article  CAS  PubMed  Google Scholar 

  49. Herbst T, Sichelstiel A, Schär C, Yadava K, Bürki K, Cahenzli J, et al. Dysregulation of allergic airway inflammation in the absence of microbial colonization. Am J Respir Crit Care Med. 2011;184:198–205. https://doi.org/10.1164/rccm.201010-1574OC.

    Article  CAS  PubMed  Google Scholar 

  50. Hill DA, Siracusa MC, Abt MC, Kim BS, Kobuley D, Kubo M, et al. Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic inflammation. Nat Med. 2012;18:538–46. https://doi.org/10.1038/nm.2657.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. •• Ohnmacht C, Park J-H, Cording S, MUCOSAL IMMUNOLOGY, et al. The microbiota regulates type 2 immunity through RORγt+ T cells. Science. 2015;349:989–93. https://doi.org/10.1126/science.aac4263. This study provides insights into the ways in which microbiota regulate type 2 immunity. RORγt+ Treg cells were profoundly reduced in germ-free or antibiotic-treated mice. In the absence of RORγt+ Treg cells, Th2-associated pathology after helminth infection was exacerbated, indicating that the microbiota regulated type 2 responses through the induction of RORγt+ Treg cells.

    Article  CAS  PubMed  Google Scholar 

  52. Geuking MB, Cahenzli J, Lawson MAE, Ng DCK, Slack E, Hapfelmeier S, et al. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity. 2011;34:794–806. https://doi.org/10.1016/j.immuni.2011.03.021.

    Article  CAS  PubMed  Google Scholar 

  53. Faith JJ, Ahern PP, Ridaura VK, Cheng J, Gordon JI. Identifying gut microbe-host phenotype relationships using combinatorial communities in gnotobiotic mice. Sci Transl Med. 2014;6:220ra11. https://doi.org/10.1126/scitranslmed.3008051.

    Article  PubMed  PubMed Central  Google Scholar 

  54. •• Mortha A, Chudnovskiy A, Hashimoto D, et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science. 2014;343:1249288. https://doi.org/10.1126/science.1249288. This key study provides mechanistic insights into the role of commensal microbes in oral tolerance. In mice, microbial sensing by intestinal macrophage promoted IL-1β secretion, leading to GM-CSF release by type 3 innate lymphoid cells, and retinoid acid and IL-10 secretion by dendritic cells and macrophages, resulting in induction and expansion of mucosal Treg cells.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139:485–98. https://doi.org/10.1016/j.cell.2009.09.033.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Atarashi K, Tanoue T, Ando M, Kamada N, Nagano Y, Narushima S, et al. Th17 cell induction by adhesion of microbes to intestinal epithelial cells. Cell. 2015;163:367–80. https://doi.org/10.1016/j.cell.2015.08.058.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. • Wang S, Charbonnier L-M, Noval Rivas M, et al. MyD88 adaptor-dependent microbial sensing by regulatory T cells promotes mucosal tolerance and enforces commensalism. Immunity. 2015;43:289–303. https://doi.org/10.1016/j.immuni.2015.06.014. In this murine study, Treg-cell-specific deletion of TLR adaptor MyD88 resulted in deficiency of intestinal Treg cell and impaired intestinal IgA production. This study indicated that microbial sensing by Treg cells via the TLR-MyD88 pathway was central to the promotion of mucosal tolerance.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Sabat R, Ouyang W, Wolk K. Therapeutic opportunities of the IL-22-IL-22R1 system. Nat Rev Drug Discov. 2014;13:21–38. https://doi.org/10.1038/nrd4176.

    Article  CAS  PubMed  Google Scholar 

  59. Sugimoto K, Ogawa A, Mizoguchi E, Shimomura Y, Andoh A, Bhan AK, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Invest. 2008;118:534–44. https://doi.org/10.1172/JCI33194.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Barcik W, Pugin B, Westermann P, Perez NR, Ferstl R, Wawrzyniak M, et al. Histamine-secreting microbes are increased in the gut of adult asthma patients. J Allergy Clin Immunol. 2016;138:1491–1494.e7. https://doi.org/10.1016/j.jaci.2016.05.049.

    Article  PubMed  Google Scholar 

  61. Ferstl R, Frei R, Schiavi E, Konieczna P, Barcik W, Ziegler M, et al. Histamine receptor 2 is a key influence in immune responses to intestinal histamine-secreting microbes. J Allergy Clin Immunol. 2014;134:744–746.e3. https://doi.org/10.1016/j.jaci.2014.04.034.

    Article  CAS  PubMed  Google Scholar 

  62. Thompson-Chagoyan OC, Vieites JM, Maldonado J, Edwards C, Gil A. Changes in faecal microbiota of infants with cow’s milk protein allergy—a Spanish prospective case-control 6-month follow-up study. Pediatr Allergy Immunol. 2010;21:e394–400. https://doi.org/10.1111/j.1399-3038.2009.00961.x.

    Article  PubMed  Google Scholar 

  63. Thompson-Chagoyan OC, Fallani M, Maldonado J, Vieites JM, Khanna S, Edwards C, et al. Faecal microbiota and short-chain fatty acid levels in faeces from infants with cow's milk protein allergy. Int Arch Allergy Immunol. 2011;156:325–32. https://doi.org/10.1159/000323893.

    Article  CAS  PubMed  Google Scholar 

  64. Nylund L, Satokari R, Nikkilä J, Rajilić-Stojanović M, Kalliomäki M, Isolauri E, et al. Microarray analysis reveals marked intestinal microbiota aberrancy in infants having eczema compared to healthy children in at-risk for atopic disease. BMC Microbiol. 2013;13:12. https://doi.org/10.1186/1471-2180-13-12.

    Article  PubMed  PubMed Central  Google Scholar 

  65. • David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–63. https://doi.org/10.1038/nature12820. This was the first study to demonstrate that the composition of gut microbiome can rapidly respond to diet. Its demonstration of close interaction between diet and commensal microbes indicates new possibilities for manipulations of the gut microbiome.

    Article  CAS  PubMed  Google Scholar 

  66. • Tang MLK, Ponsonby A-L, Orsini F, et al. Administration of a probiotic with peanut oral immunotherapy: a randomized trial. J Allergy Clin Immunol. 2015;135:737–44.e8. https://doi.org/10.1016/j.jaci.2014.11.034. This was the first placebo-controlled randomized trial in peanut-allergic children utilizing probiotic ( Lactobacillus rhamnosus ) and peanut oral immunotherapy. High sustained unresponsiveness to peanut (82.1%) was reported in the treatment arm; however, the specific contribution of probiotic was difficult to determine because there was no oral immunotherapy-only arm.

    Article  CAS  PubMed  Google Scholar 

  67. Tan J, McKenzie C, Vuillermin PJ, Goverse G, Vinuesa CG, Mebius RE, et al. Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways. Cell Rep. 2016;15:2809–24. https://doi.org/10.1016/j.celrep.2016.05.047.

    Article  CAS  PubMed  Google Scholar 

  68. Böttcher MF, Nordin EK, Sandin A, Midtvedt T, Björkstén B. Microflora-associated characteristics in faeces from allergic and nonallergic infants. Clin Exp Allergy. 2000;30:1590–6.

    Article  PubMed  Google Scholar 

  69. Huffnagle GB, Noverr MC. The emerging world of the fungal microbiome. Trends Microbiol. 2013;21:334–41. https://doi.org/10.1016/j.tim.2013.04.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Zou S, Caler L, Colombini-Hatch S, Glynn S, Srinivas P. Research on the human virome: where are we and what is next. Microbiome. 2016;4:32. https://doi.org/10.1186/s40168-016-0177-y.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Waldor MK, Tyson G, Borenstein E, Ochman H, Moeller A, Finlay BB, et al. Where next for microbiome research? PLoS Biol. 2015;13:e1002050. https://doi.org/10.1371/journal.pbio.1002050.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Cadwell K. The virome in host health and disease. Immunity. 2015;42:805–13. https://doi.org/10.1016/j.immuni.2015.05.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. • Bunyavanich S, Schadt EE. Systems biology of asthma and allergic diseases: a multiscale approach. J Allergy Clin Immunol. 2015;135:31–42. https://doi.org/10.1016/j.jaci.2014.10.015. A systems biology approach to asthma and allergic diseases could enable a more comprehensive understanding of the microbiome and complex disorders. Here the authors review recent applications of system-wide profiling to asthma and allergy and provide perspective on building network models to integrate multiscale data

    Article  PubMed  Google Scholar 

  74. Watson CT, Cohain AT, Griffin RS, Chun Y, Grishin A, Hacyznska H, et al. Integrative transcriptomic analysis reveals key drivers of acute peanut allergic reactions. Nat Commun. 2017;8:1943. https://doi.org/10.1038/s41467-017-02188-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Chen Y, Zhu J, Lum PY, Yang X, Pinto S, MacNeil DJ, et al. Variations in DNA elucidate molecular networks that cause disease. Nature. 2008;452:429–35. https://doi.org/10.1038/nature06757.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Miller CL, Pjanic M, Wang T, Nguyen T, Cohain A, Lee JD, et al. Integrative functional genomics identifies regulatory mechanisms at coronary artery disease loci. Nat Commun. 2016;7:12092. https://doi.org/10.1038/ncomms12092.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Sonnenburg JL, Bäckhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535:56–64. https://doi.org/10.1038/nature18846.

    Article  CAS  PubMed  Google Scholar 

  78. Neef A, Sanz Y. Future for probiotic science in functional food and dietary supplement development. Curr Opin Clin Nutr Metab Care. 2013;16:679–87. https://doi.org/10.1097/MCO.0b013e328365c258.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Supinda Bunyavanich.

Ethics declarations

Conflict of Interest

The authors declare no conflicts of interest relevant to this manuscript.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

This article is part of the Topical Collection on Food Allergy

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ho, He., Bunyavanich, S. Role of the Microbiome in Food Allergy. Curr Allergy Asthma Rep 18, 27 (2018). https://doi.org/10.1007/s11882-018-0780-z

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11882-018-0780-z

Keywords

Navigation