Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Microbiota therapy acts via a regulatory T cell MyD88/RORγt pathway to suppress food allergy

An Author Correction to this article was published on 16 August 2019

This article has been updated

Abstract

The role of dysbiosis in food allergy (FA) remains unclear. We found that dysbiotic fecal microbiota in FA infants evolved compositionally over time and failed to protect against FA in mice. Infants and mice with FA had decreased IgA and increased IgE binding to fecal bacteria, indicative of a broader breakdown of oral tolerance than hitherto appreciated. Therapy with Clostridiales species impacted by dysbiosis, either as a consortium or as monotherapy with Subdoligranulum variabile, suppressed FA in mice as did a separate immunomodulatory Bacteroidales consortium. Bacteriotherapy induced expression by regulatory T (Treg) cells of the transcription factor ROR-γt in a MyD88-dependent manner, which was deficient in FA infants and mice and ineffectively induced by their microbiota. Deletion of Myd88 or Rorc in Treg cells abrogated protection by bacteriotherapy. Thus, commensals activate a MyD88/ROR-γt pathway in nascent Treg cells to protect against FA, while dysbiosis impairs this regulatory response to promote disease.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Infants with FA exhibit an evolving gut dysbiosis.
Fig. 2: Altered mucosal antibody responses to the gut microbiota in FA.
Fig. 3: A consortium of Clostridiales species prevents FA.
Fig. 4: Clostridiales and Bacteroidales consortia suppress established FA.
Fig. 5: ROR-γt+ Treg cell deficiency promotes FA.
Fig. 6: Protection against FA by commensals requires ROR-γt+ Treg cells.

Data availability

Data presented in the manuscript, including de-identified patient results, will be made available to investigators following request. All requests for raw and analyzed data and materials will be promptly reviewed by the Boston Children’s Hospital Technology & Innovation Development Office to verify whether the request is subject to any intellectual property or confidentiality obligations. Any data and materials that can be shared will be released via a Material Transfer Agreement. The 16S bacterial rRNA datasets generated in the course of this project have been deposited at the National Center for Biotechnology Information Sequence Read Archive under BioProject ID: PRJNA525231. Detailed age and disease attributes of the deposited BioProject sample data will be made available on request.

Change history

  • 16 August 2019

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Branum, A. M. & Lukacs, S. L. Food allergy among children in the United States. Pediatrics 124, 1549–1555 (2009).

    PubMed  Google Scholar 

  2. Wills-Karp, M., Santeliz, J. & Karp, C. L. The germless theory of allergic disease: revisiting the hygiene hypothesis. Nat. Rev. Immunol. 1, 69–75 (2001).

    CAS  PubMed  Google Scholar 

  3. Ly, N. P., Litonjua, A., Gold, D. R. & Celedon, J. C. Gut microbiota, probiotics, and vitamin D: interrelated exposures influencing allergy, asthma, and obesity? J. Allergy Clin. Immunol. 127, 1087–1094 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Koplin, J. et al. Is caesarean delivery associated with sensitization to food allergens and IgE-mediated food allergy? A systematic review. Pediatr. Allergy Immunol. 19, 682–687 (2008).

    PubMed  Google Scholar 

  5. Bjorksten, B. Disease outcomes as a consequence of environmental influences on the development of the immune system. Curr. Opin. Allergy Clin. Immunol. 9, 185–189 (2009).

    PubMed  Google Scholar 

  6. Rachid, R. & Chatila, T. A. The role of the gut microbiota in food allergy. Curr. Opin. Pediatr. 28, 748–753 (2016).

    CAS  PubMed  Google Scholar 

  7. Azad, M. B. et al. Infant gut microbiota and food sensitization: associations in the first year of life. Clin. Exp. Allergy 45, 632–643 (2015).

    CAS  PubMed  Google Scholar 

  8. Sudo, N. et al. The requirement of intestinal bacterial flora for the development of an IgE production system fully susceptible to oral tolerance induction. J. Immunol. 159, 1739–1745 (1997).

    CAS  PubMed  Google Scholar 

  9. Fritz, J. H. et al. Acquisition of a multifunctional IgA+plasma cell phenotype in the gut. Nature 481, 199–203 (2012).

    CAS  Google Scholar 

  10. Geuking, M. B. et al. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity 34, 794–806 (2011).

    CAS  PubMed  Google Scholar 

  11. Stefka, A. T. et al. Commensal bacteria protect against food allergen sensitization. Proc. Natl Acad. Sci. USA 111, 13145–13150 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Atarashi, K. et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337–341 (2011).

    CAS  PubMed  Google Scholar 

  13. Noval Rivas, M. et al. A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis. J. Allergy Clin. Immunol. 131, 201–212 (2013).

    CAS  PubMed  Google Scholar 

  14. Wesemann, D. R. & Nagler, C. R. The microbiome, timing, and barrier function in the context of allergic disease. Immunity 44, 728–738 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Noval Rivas, M. & Chatila, T. A. Regulatory T cells in allergic diseases. J. Allergy Clin. Immunol. 138, 639–652 (2016).

    CAS  PubMed  Google Scholar 

  16. Abdel-Gadir, A., Massoud, A. H. & Chatila, T. A. Antigen-specific Treg cells in immunological tolerance: implications for allergic diseases. F1000Res. 7, 38 (2018).

    PubMed  PubMed Central  Google Scholar 

  17. Furusawa, Y. et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504, 446–450 (2013).

    CAS  PubMed  Google Scholar 

  18. Smith, P. M. et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573 (2013).

    CAS  PubMed  Google Scholar 

  19. Arpaia, N. et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504, 451–455 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Tan, J. et al. Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways. Cell Rep. 15, 2809–2824 (2016).

    CAS  PubMed  Google Scholar 

  21. Ohnmacht, C. et al. MUCOSAL IMMUNOLOGY. The microbiota regulates type 2 immunity through RORγt +T cells. Science 349, 989–993 (2015).

    CAS  PubMed  Google Scholar 

  22. Sefik, E. et al. MUCOSAL IMMUNOLOGY. Individual intestinal symbionts induce a distinct population of RORγ+ regulatory T cells. Science 349, 993–997 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Burton, O. T. et al. Immunoglobulin E signal inhibition during allergen ingestion leads to reversal of established food allergy and induction of regulatory T cells. Immunity 41, 141–151 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Noval Rivas, M., Burton, O. T., Oettgen, H. C. & Chatila, T. IL-4 production by group 2 innate lymphoid cells promotes food allergy by blocking regulatory T-cell function. J. Allergy Clin. Immunol. 138, 801–811.e9 (2016).

    CAS  PubMed  Google Scholar 

  25. Noval Rivas, M. et al. Regulatory T cell reprogramming toward a Th2-cell-like lineage impairs oral tolerance and promotes food allergy. Immunity 42, 512–523 (2015).

    CAS  PubMed  Google Scholar 

  26. Fagarasan, S. et al. Critical roles of activation-induced cytidine deaminase in the homeostasis of gut flora. Science 298, 1424–1427 (2002).

    CAS  PubMed  Google Scholar 

  27. Kubinak, J. L. et al. MyD88 signaling in T cells directs IgA-mediated control of the microbiota to promote health. Cell Host Microbe 17, 153–163 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Wang, S. et al. MyD88 adaptor-dependent microbial sensing by regulatory T cells promotes mucosal tolerance and enforces commensalism. Immunity 43, 289–303 (2015).

    PubMed  PubMed Central  Google Scholar 

  29. Donaldson, G. P. et al. Gut microbiota utilize immunoglobulin A for mucosal colonization. Science 360, 795–800 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Macpherson, A. J., Yilmaz, B., Limenitakis, J. P. & Ganal-Vonarburg, S. C. IgA function in relation to the intestinal microbiota. Annu. Rev. Immunol. 36, 359–381 (2018).

    CAS  PubMed  Google Scholar 

  31. Mathias, C. B. et al. IgE-mediated systemic anaphylaxis and impaired tolerance to food antigens in mice with enhanced IL-4 receptor signaling. J. Allergy Clin. Immunol. 127, e791–e796 (2011).

    Google Scholar 

  32. Kalia, V. C. et al. Analysis of the unexplored features of rrs (16S rDNA) of the genus Clostridium. BMC Genom. 12, 18 (2011).

    CAS  Google Scholar 

  33. Rajilic-Stojanovic, M. & de Vos, W. M. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol. Rev. 38, 996–1047 (2014).

    CAS  PubMed  Google Scholar 

  34. Blander, J. M., Longman, R. S., Iliev, I. D., Sonnenberg, G. F. & Artis, D. Regulation of inflammation by microbiota interactions with the host. Nat. Immunol. 18, 851–860 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Narushima, S. et al. Characterization of the 17 strains of regulatory T cell-inducing human-derived Clostridia. Gut Microbes 5, 333–339 (2014).

    PubMed  PubMed Central  Google Scholar 

  36. Walker, A. W. & Lawley, T. D. Therapeutic modulation of intestinal dysbiosis. Pharm. Res. 69, 75–86 (2013).

    CAS  Google Scholar 

  37. Kim, K. S. et al. Dietary antigens limit mucosal immunity by inducing regulatory T cells in the small intestine. Science 351, 858–863 (2016).

    CAS  PubMed  Google Scholar 

  38. Lathrop, S. K. et al. Peripheral education of the immune system by colonic commensal microbiota. Nature 478, 250–254 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Russler-Germain, E. V., Rengarajan, S. & Hsieh, C. S. Antigen-specific regulatory T-cell responses to intestinal microbiota. Mucosal Immunol. 10, 1375–1386 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Thornton, A. M. et al. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3. +T regulatory cells. J. Immunol. 184, 3433–3441 (2010).

    CAS  PubMed  Google Scholar 

  41. Weiss, J. M. et al. Neuropilin 1 is expressed on thymus-derived natural regulatory T cells, but not mucosa-generated induced Foxp3. +T reg cells. J. Exp. Med. 209, S1721 (2012).

    Google Scholar 

  42. Yadav, M. et al. Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo. J. Exp. Med. 209, S1711–S1719 (2012).

    Google Scholar 

  43. Bilate, A. M. & Lafaille, J. J. Induced CD4 +Foxp3+ regulatory T cells in immune tolerance. Annu Rev. Immunol. 30, 733–758 (2012).

    CAS  PubMed  Google Scholar 

  44. Xu, M. et al. c-MAF-dependent regulatory T cells mediate immunological tolerance to a gut pathobiont. Nature 554, 373–377 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Abdel-Gadir, A. et al. Oral immunotherapy with omalizumab reverses the Th2 cell-like programme of regulatory T cells and restores their function. Clin. Exp. Allergy 48, 825–836 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Bartnikas, L. M. et al. Epicutaneous sensitization results in IgE-dependent intestinal mast cell expansion and food-induced anaphylaxis. J. Allergy Clin. Immunol. 131, e451–e456 (2013).

    Google Scholar 

  47. Round, J. L. et al. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 332, 974–977 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Kverka, M. et al. Oral administration of Parabacteroides distasonis antigens attenuates experimental murine colitis through modulation of immunity and microbiota composition. Clin. Exp. Immunol. 163, 250–259 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Geva-Zatorsky, N. et al. Mining the human gut microbiota for immunomodulatory organisms. Cell 168, 928–943 e911 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Mangalam, A. et al. Human gut-derived commensal bacteria suppress CNS inflammatory and demyelinating disease. Cell Rep. 20, 1269–1277 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Haribhai, D. et al. A requisite role for induced regulatory T cells in tolerance based on expanding antigen receptor diversity. Immunity 35, 109–122 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Rios-Covian, D., Salazar, N., Gueimonde, M. & de Los Reyes-Gavilan, C. G. Shaping the metabolism of intestinal Bacteroides population through diet to improve human health. Front. Microbiol. 8, 376 (2017).

    PubMed  PubMed Central  Google Scholar 

  53. Atarashi, K. et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 500, 232–236 (2013).

    CAS  PubMed  Google Scholar 

  54. Britton, G. J. et al. Microbiotas from humans with inflammatory bowel disease alter the balance of gut Th17 and RORγt +regulatory T cells and exacerbate colitis in mice.Immunity 50, 212–224 e214 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Dethlefsen, L. & Relman, D. A. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc. Natl Acad. Sci. USA 108 (Suppl. 1), 4554–4561 (2011).

    CAS  PubMed  Google Scholar 

  56. Gerber, G. K., Onderdonk, A. B. & Bry, L. Inferring dynamic signatures of microbes in complex host ecosystems. PLoS Comput. Biol. 8, e1002624 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Bucci, V. et al. MDSINE: microbial dynamical systems inference engine for microbiome time-series analyses. Genome Biol. 17, 121 (2016).

    PubMed  PubMed Central  Google Scholar 

  58. Schloss, P. D. et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 75, 7537–7541 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Excoffier, L., Smouse, P. E. & Quattro, J. M. Analysis of molecular variance inferred from metric distances among DNA haplotypes: application to human mitochondrial DNA restriction data. Genetics 131, 479–491 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Lozupone, C., Hamady, M. & Knight, R. UniFrac–an online tool for comparing microbial community diversity in a phylogenetic context. BMC Bioinforma. 7, 371 (2006).

    Google Scholar 

  61. Lozupone, C. & Knight, R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl. Environ. Microbiol. 71, 8228–8235 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. McMurdie, P. & Holmes, S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE 8, e61217 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. McMurdie, P. & Holmes, S. Waste not, want not: why rarefying microbiome data is inadmissible. PLoS Comput. Biol. 10, e1003531 (2014).

    PubMed  PubMed Central  Google Scholar 

  64. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B 57, 289–300 (1995).

    Google Scholar 

  65. Matsen, F., Kodner, R. B. & Armbrust, E. V. pplacer: linear time maximum-likelihood and Bayesian phylogenetic placement of sequences onto a fixed reference tree. BMC Bioinforma. 11, 538 (2010).

    Google Scholar 

  66. Cole, J. R. et al. Ribosomal Database Project: data and tools for high throughput rRNA analysis. Nucleic acids Res. 42, D633–642 (2014).

    CAS  PubMed  Google Scholar 

  67. Massoud, A. H. et al. An asthma-associated IL4R variant exacerbates airway inflammation by promoting conversion of regulatory T cells to TH17-like cells. Nat. Med. 22, 1013–1022 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Rubtsov, Y. P. et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546–558 (2008).

    CAS  PubMed  Google Scholar 

  69. Pinciroli, R. et al. Endotracheal Tubes cleaned with a novel mechanism for secretion removal: a randomized controlled clinical study. Respir. Care 61, 1431–1439 (2016).

    PubMed  Google Scholar 

Download references

Acknowledgements

We thank H. Oettgen for the provision of Igh7–/–Il4raF709 mice, L.-M. Charbonnier for critical review of the manuscript and M. Delaney for support with microbiology. This work was supported by NIH NIAID grants Nos. 1R56AI117983 and 1R01AI126915 (to T.A.C.), NIDDK grant No. P30DK056338 (to L.B.), the Clinical and Translational Science Center/Harvard Catalyst, the Bunning Food Allergy Fund, the Jasmine and Paul Mashikian Fund, the Massachusetts Life Sciences Center and a Partners Healthcare Innovations Development grant.

Author information

Authors and Affiliations

Authors

Contributions

T.A.C., R.R. and D.T.U. conceived the human microbiota studies, and T.A.C. conceived the mechanistic studies and directed the overall project. L.B. conceived the bacterial consortia and oversaw their development for use as a therapeutic. G.K.G., N.L. and X.D. carried out the bioinformatic analyses of human fecal microbiota composition. N.D. designed multiplex probes for the consortia and carried out the persistence studies. A.A.-G., E.S.-V., M.N.R., S.W., H.H. and L.W. carried out the experiments and evaluated the data. R.R. oversaw the design and execution of the human studies. S.S., W.S., E.C. and H.B. were involved in human subject recruitment and/or the collection of fecal samples. T.A.C. and A.A.-G. wrote the manuscript.

Corresponding authors

Correspondence to Rima Rachid or Talal A. Chatila.

Ethics declarations

Competing interests

L.B., G.K.G., T.A.C., R.R. and A.A.-G. are inventors on published US patent application No. 15/801,811, submitted by The Brigham and Women’s Hospital, Inc. and Children’s Medical Center Corporation, that covers methods and compositions for the prevention and treatment of food allergy using microbial treatments. T.A.C., R.R., A.A.-G. and E.S.-V. have pending patent applications related to the use of probiotics in enforcing oral tolerance in food allergy (Nos. 62/758, 161, and 62/823,866). L.B., G.K.G. and T.A.C. are founders of, and have equity in, Consortia Tx. R.R. has equity in Consortia Tx. A.A.-G. is currently an employee of, and owns shares in, Seed Health Inc. The rest of the authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 FMT from WT mice protects against FA in GF Il4raF709 mice.

a, Temperature changes in GF Il4raF709 mice that were either left uncolonized or reconstituted with FMT from WT or Il4raF709 mice, then sensitized with OVA/SEB and challenged with OVA (n = 15 WT and 14 Il4raF709 mice). b,c, Total and OVA-specific serum IgE (n = 15 WT and 14 Il4raF709 mice). d, MMCP-1 concentrations post-OVA challenge (n = 6 per group). e,f, Analysis of ROR-γt and GATA3 expression in MLN HeliosNRP1 and Helios+NRP1+ Treg cells (n = 6 per group). Each dot represents one mouse. Data represent mean ± s.e.m. from two or three independent experiments. P values were derived by repeat measures two-way ANOVA (a) or by Student’s unpaired two-tailed t-test with Welch correction (bf).

Source data

Extended Data Fig. 2 Analysis of IgA- and IgE-bound bacteria in fecal samples.

a,c, Representative FACS plots showing the gating strategy for human (a) and mouse (c) fecal bacteria. Bacteria present in the feces were identified by gating on SYTO-BC+ events (right-side panels). b,d, Frequencies of IgA- and IgE-bound bacteria as assessed by gating on bacteria bound with the respective PE-labeled anti-IgA and anti-IgE antibodies, as shown in a and c. e,f, Analysis of sIgA+ (e) and IgE+ (f) fecal bacteria from Il4raF709 mice sensitized with OVA/SEB without or with bacterial therapy. Fecal pellets from Rag2–/– and Igh7–/–Il4raF709 mice were used as negative controls. Each symbol in the scatter plots represents one mouse (no treatment: n = 11 per group; Clostridiales: n = 8 per group; Proteobacteria: n = 9 and 7). Data represent mean ± s.e.m. from two independent experiments. Flow panels in c,d are representative of two two independent experiments. P values were derived by one-way ANOVA with Dunnett’s post hoc analysis.

Source data

Extended Data Fig. 3 Antibiotic therapy potentiates the therapeutic efficacy of the Clostridiales consortium in Il4raF709 mice.

a, Temperature changes in the respective OVA/SEB-sensitized and OVA-challenged SPF Il4raF709 mouse groups treated as follows: no antibiotics (n = 6), Clostridiales (n = 5) and antibiotics without or with Clostridiales (n = 5 per group). P values were derived by two-way ANOVA. b,c, Total and OVA-specific IgE: no antibiotics (n = 4 per group), Clostridiales (n = 5 per group) and antibiotics without (n = 5 per group) or with Clostridiales (n = 4 per group). d, MMCP-1 concentrations: no antibiotics (n = 5), Clostridiales (n = 5) and antibiotics without (n = 4) or with Clostridiales (n = 4). e, Frequencies of total CD4+Foxp3+, HeliosNRP1Foxp3+, ROR-γt+CD4+Foxp3+ and IL-4+ CD4+Foxp3+ Treg cells in the MLN of the respective mouse group: no antibiotics (n = 6), Clostridiales (n = 5) and antibiotics without (n = 5) or with Clostridiales (n = 5). Each dot represents one mouse. Throughout, data represent mean ± s.e.m. from two independent experiments. Unless otherwise indicated, P values were derived by one-way ANOVA with Dunnett’s post hoc analysis.

Source data

Extended Data Fig. 4 Bacteriotherapy with S. variabile protects against FA.

a, Temperature changes in SPF Il4raF709 mice that were antibiotic-treated then sensitized with OVA/SEB while receiving no treatment (n = 8) or treatment with S. variabile (n = 11), and thereafter challenged with OVA. P values were derived by two-way ANOVA. b,c, Total and OVA-specific IgE (no bacteria: n = 8; S. variabile: n = 11). d, MMCP-1 concentrations (no bacteria: n = 8; S. variabile: n = 11). e,f, Analysis of MLN ROR-γt+ and GATA3+ cells among HeliosNRP1 and Helios+NRP1+ Foxp3+ Treg cells, respectively (no bacteria: n = 8; S. variabile: n = 5). g, Analysis of MLN IL-4+ CD4+Foxp3+ Treg cells and IL-4+ CD4+Foxp3 Teff cells (no bacteria: n = 8; S. variabile: n = 5). Each dot represents one mouse. Throughout, data represent mean ± s.e.m. from two independent experiments. For bg, P values were derived by Student’s unpaired two-tailed t-test with Welch correction.

Source data

Extended Data Fig. 5 Clostridiales protects against percutaneous sensitization-induced FA.

a, Temperature changes in SPF WT BALB/c mice that were antibiotic-treated then percutaneously sensitized with OVA/SEB while receiving either no treatment (n = 14) or treatment with Clostridales (n = 11), and thereafter challenged with OVA. P values were derived by two-way ANOVA. b,c, Total and OVA-specific IgE concentrations (n = 7 per group). d, MMCP-1 concentrations (n = 7 per group). e, Analysis of small intestinal LPL ROR-γt+ CD4+Foxp3+ Treg cells (n = 7 per group). Each dot represents one mouse. Data represent mean ± s.e.m. from two independent experiments. For be, P values were derived by Student’s unpaired two-tailed t-test with Welch correction.

Source data

Extended Data Fig. 6 A Bacteroidales consortium prevents FA.

a, Left: experimental schema; right: temperature changes in GF Il4raF709 mice that were colonized and sensitized as indicated then challenged with OVA (n = 5 per group). b,c, Total and OVA-specific IgE (b) and MMCP-1 concentrations (c). GF, OVA/SEB (n = 5 per group), Bacteroidales, PBS (n = 6, 7 and 7), Bacteroidales, OVA/SEB (n = 6, 6 and 7). d, Frequencies of MLN CD4+Foxp3+, IL-4+Foxp3+ and GATA3+Foxp3+ T cells. GF, OVA/SEB (n = 5 per group), Bacteroidales, PBS (n = 4, 8 and 5), Bacteroidales, OVA/SEB (n = 6, 5 and 6). e, Frequencies of HeliosNrp1Foxp3+ and ROR-γt+Foxp3+ T cells. GF, OVA/SEB (n = 5 and 7), Bacteroidales, PBS (n = 5 per group), Bacteroidales, OVA/SEB (n = 6 per group). f, Left: experimental schema; right: temperature changes in Il4raF709 mice sensitized and treated as indicated. OVA/SEB (n = 6), OVA/SEB, Bacteroidales, (n = 5). g, Total and OVA-specific IgE and MMCP-1 concentrations: OVA/SEB (n = 7, 9 and 9), OVA/SEB, Bacteroidales, (n = 5, 10 and 5). h,i, Frequencies of MLN CD4+Foxp3+, IL-4+Foxp3+ and GATA3+Foxp3+ (h) and HeliosNrp1Foxp3+ and ROR-γt+Foxp3+ T cells (i). OVA/SEB (n = 5, 8 and 3, 5 and 5), OVA/SEB, Bacteroidales (n = 5, 10, 4, 6 and 9). j, IgE and IgA staining of fecal bacteria. OVA/SEB (n = 11 per group), OVA/SEB, Bacteroidales (n = 8 per group). Each dot represents one mouse. Data represent mean ± s.e.m. from two independent experiments. P values were derived by repeat measures two-way ANOVA (a,f), by one-way ANOVA with Dunnett’s post hoc analysis (be) or by Student’s unpaired two-tailed t-test (hj).

Source data

Extended Data Fig. 7 Depletion of Treg cells abrogates protection by the microbiota.

a, Experimental schema. b, Temperature changes in the respective OVA/SEB-sensitized and OVA-challenged mouse groups: Il4raF709Foxp3EGFP/DTR– (n = 6), Il4raF709Foxp3EGFP/DTR–+Clostridiales+DT (n = 7), Il4raF709Foxp3EGFP/DTR++Clostridiales+DT (n = 8), Il4raF709Foxp3EGFP/DTR–+Bacteroidales+DT (n = 9), Il4raF709Foxp3EGFP/DTR++Bacteroidales+DT (n = 7). c, Total and OVA-specific IgE in the groups listed in b (total IgE: n = 9, 6, 8, 7 and 5; OVA-specific IgE: 6, 5, 5, 5 and 5). d, MMCP-1 concentrations (n = 12 for Il4raF709Foxp3EGFP/DTR–, and n = 8 per group for all other groups). e,f, Frequencies of MLN CD4+Foxp3+ and IL-4+Foxp3+ T cells: Il4raF709Foxp3EGFP/DTR– (n = 6 and 10), Il4raF709Foxp3EGFP/DTR–+Clostridiales+DT (n = 8 and 7), Il4raF709Foxp3EGFP/DTR++Clostridiales+DT (n = 8 and 7), Il4raF709Foxp3EGFP/DTR–+Bacteroidales+DT (n = 8 and 7), Il4raF709Foxp3EGFP/DTR++Bacteroidales+DT (n = 8 and 7). g, Frequencies of ROR-γt+Foxp3+ and GATA3+Foxp3+ T cells: Il4raF709Foxp3EGFP/DTR– (n = 5 per group), Il4raF709Foxp3EGFP/DTR–+Clostridiales+DT (n = 7 and 9), Il4raF709Foxp3EGFP/DTR++Clostridiales+DT (n = 8 per group), Il4raF709Foxp3EGFP/DTR–+Bacteroidales+DT (n = 6 per group), Il4raF709Foxp3EGFP/DTR++Bacteroidales+DT (n = 8 pre group). Each dot represents one mouse. Data represent mean ± s.e.m. from two independent experiments. P values were derived by repeat measures two-way ANOVA (b) or by one-way ANOVA with Dunnett’s post hoc analysis or Student’s unpaired two-tailed t-test (cf).

Source data

Extended Data Fig. 8 Oral SCFA supplementation does not protect against FA.

a, SCFA in fecal samples of PBS or OVA/SEB-sensitized WT and Il4raF709 mice. Acetate, propionate and butyrate: WT, PBS or OVA/SEB (n = 5 per group); Il4raF709, PBS (n = 5 per group) or OVA/SEB (n = 10 per group). Isovalerate: WT, PBS or OVA/SEB (n = 5 and 4); Il4raF709, PBS or OVA/SEB (n = 5 and 8). Valerate: WT, PBS or OVA/SEB (n = 5 and 3); Il4raF709, PBS or OVA/SEB (n = 4 and 7). b, Temperature changes in OVA-challenged WT and Il4raF709 mice sensitized with PBS or OVA/SEB without or with SCFA supplementation: WT, PBS+SCFA (n = 10), WT, OVA/SEB (n = 11), WT, OVA/SEB+SCFA (n = 24); Il4raF709, PBS+SCFA (n = 12), Il4raF709, OVA/SEB (n = 7), Il4raF709, OVA/SEB+SCFA (n = 17). c, Total and OVA-specific IgE: WT, PBS+SCFA (n = 5 per group), WT, OVA/SEB (n = 6 per group), WT, OVA/SEB+SCFA (n = 9 per group); Il4raF709, PBS+SCFA (n = 4 per group), Il4raF709, OVA/SEB (n = 8 per group), Il4raF709, OVA/SEB+SCFA (n = 9 per group). d, Frequencies of MLN CD4+Foxp3+ROR-γt+ and CD4+Foxp3ROR-γt+ T cells: WT, PBS+SCFA (n = 5 per group), WT, OVA/SEB (n = 4 per group), WT, OVA/SEB+SCFA (n = 4 per group); Il4raF709, PBS+SCFA (n = 5 per group), Il4raF709, OVA/SEB (n = 5 per group), Il4raF709, OVA/SEB+SCFA (n = 7 per group). Each dot represents one mouse. Data represent mean ± s.e.m. from two independent experiments. P values were derived by the Kolmogorov–Smirnov test (a), by Student’s unpaired two-tailed t-test (c,d) or by two-way ANOVA (b).

Source data

Extended Data Fig. 9 Analysis of ROR-γt+ expression in human subjects and mutant mice.

a, Gating strategy for CD4+Foxp3+ (G1) and CD4+Foxp3 T (G2) cells ex vivo. b, Gating strategy for the expression of ROR-γt in Teff cells (G2) from patients with FA, healthy controls (HC) and atopic subjects (atopy), as compared to an isotype control. c, Flow plots and frequencies of peripheral blood CD4+Foxp3+ROR-γt+ T cells in WT and Il4raF709 mice (n = 7 mice per group). d, Flow plots and frequencies of peripheral blood CD4+Foxp3+HeliosNRP1ROR-γt+ T cells in WT and Il4raF709 mice (n = 7 mice per group). e,f, Flow plots and frequencies of MLN CD4+Foxp3+ROR-γt+ T cells from Foxp3YFPCre mice sensitized with OVA/SEB, and Foxp3YFPCreRorcΔ/Δ either sham-sensitized (PBS) or sensitized with OVA/SEB, as indicated (n = 5 mice per group). g, Quantitative PCR with reverse transcription of Rorc gene expression in MLN CD4+Foxp3+ Treg and CD4+Foxp3 Teff cells from Foxp3YFPCre, Foxp3YFPCreRorcΔ/Δ and Il4raF709Foxp3YFPCreRorcΔ/Δ mice. Data were normalized to the endogenous Hprt transcripts (n = 5 mice per group). Each dot represents one mouse. Results represent means ± s.e.m. collated from two independent experiments. P values were derived by Student’s unpaired two-tailed t-test with Welch correction (c,d), or by one-way ANOVA with Dunnett’s post hoc analysis (f,g).

Source data

Extended Data Fig. 10 Treg cell-specific deletion of Rorc and Myd88 impairs mucosal tolerance.

ad. Analysis of sIgA+ (a,b) and IgE+ (c,d) fecal bacteria in OVA/SEB-sensitized Foxp3YFPCre, Il4raF709Foxp3YFPCre and Foxp3YFPCreRorcΔ/Δ mice. Fecal pellets from Rag2–/– and Igh7–/–Il4raF709 mice were used as negative controls: Foxp3YFPCre (n = 6 per group), Il4raF709Foxp3YFPCre (n = 11 and 7) and Foxp3YFPCreRorcΔ/Δ mice (n = 10 and 8). ef, Analysis of GATA3+Foxp3+ Treg cells in the following OVA/SEB-sensitized mice that were either untreated or treated with Clostridiales or Bacteroidales consortium: Il4raF709Foxp3YFPCre (n = 9, 5 and 5) and Il4raF709Foxp3YFPCreRorcΔ/Δ (n = 4, 5 and 8). g,h, Analysis of GATA3+Foxp3+ Treg cells in OVA/SEB-sensitized Il4raF709Foxp3YFPCre mice treated with the Bacteroidales consortium (n = 9), and in OVA/SEB-sensitized Il4raF709Foxp3YFPCreMyd88Δ/Δ mice otherwise untreated or treated with the Clostridiales or Bacteroidales consortium (n = 8, 9 and 8). Each symbol represents one mouse. Results represent means ± s.e.m. collated from two independent experiments. P values were derived by one-way ANOVA with Dunnett’s post hoc analysis (b,d,f,h).

Source data

Supplementary information

Supplementary information

Supplementary Methods, Supplementary Figs. 1–9 and their figure legends, Supplementary Tables 1–7 and their table legends.

Reporting Summary

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig. 8

Statistical Source Data

Source Data Extended Data Fig. 9

Statistical Source Data

Source Data Extended Data Fig. 10

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Abdel-Gadir, A., Stephen-Victor, E., Gerber, G.K. et al. Microbiota therapy acts via a regulatory T cell MyD88/RORγt pathway to suppress food allergy. Nat Med 25, 1164–1174 (2019). https://doi.org/10.1038/s41591-019-0461-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-019-0461-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing