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.

  • Article
  • Published:

Inulin-gel-based oral immunotherapy remodels the small intestinal microbiome and suppresses food allergy

Abstract

Despite the potential of oral immunotherapy against food allergy, adverse reactions and loss of desensitization hinder its clinical uptake. Dysbiosis of the gut microbiota is implicated in the increasing prevalence of food allergy, which will need to be regulated to enable for an effective oral immunotherapy against food allergy. Here we report an inulin gel formulated with an allergen that normalizes the dysregulated ileal microbiota and metabolites in allergic mice, establishes allergen-specific oral tolerance and achieves robust oral immunotherapy efficacy with sustained unresponsiveness in food allergy models. These positive outcomes are associated with enhanced allergen uptake by antigen-sampling dendritic cells in the small intestine, suppressed pathogenic type 2 immune responses, increased interferon-γ+ and interleukin-10+ regulatory T cell populations, and restored ileal abundances of Eggerthellaceae and Enterorhabdus in allergic mice. Overall, our findings underscore the therapeutic potential of the engineered allergen gel as a suitable microbiome-modulating platform for food allergy and other allergic diseases.

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

Access options

Buy this article

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

Fig. 1: Inulin gel/OVA protects mice against repeated allergen challenges with chicken egg white.
Fig. 2: Inulin gel increases the residence time and uptake of allergen in the small intestine.
Fig. 3: Inulin gel/OVA induces immune regulatory phenotype in the small intestine.
Fig. 4: Inulin gel/OVA establishes durable protection with sustained unresponsiveness.
Fig. 5: Inulin gel/OVA normalizes the dysbiotic ileal microbiota in food allergy.
Fig. 6: Therapeutic efficacy of inulin gel/allergen OIT in various food allergy models.

Similar content being viewed by others

Data availability

The scRNA data have been deposited in the NCBI Sequence Read Archive (accession no. PRJNA1074271). The bacterial 16S rRNA-sequencing data have been deposited in the NCBI Sequence Read Archive (accession no. PRJNA1073670). The data supporting the findings of this study are available within the Article and its Supplementary Information files. All relevant data are available from the corresponding author. Source data are provided with this paper.

References

  1. Sicherer, S. H. Food allergy. Lancet 360, 701–710 (2002).

    Article  PubMed  Google Scholar 

  2. Mullard, A. FDA approves first peanut allergy drug. Nat. Rev. Drug Discov. 19, 156 (2020).

    PubMed  Google Scholar 

  3. Brown, K. R. et al. Safety of peanut (Arachis hypogaea) allergen powder-dnfp in children and teenagers with peanut allergy: pooled summary of phase 3 and extension trials. J. Allergy Clin. Immunol. 149, 2043–2052. e2049 (2022).

    Article  CAS  PubMed  Google Scholar 

  4. Chinthrajah, R. S. et al. Sustained outcomes in oral immunotherapy for peanut allergy (POISED study): a large, randomised, double-blind, placebo-controlled, phase 2 study. Lancet 394, 1437–1449 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Stephen-Victor, E., Crestani, E. & Chatila, T. A. Dietary and microbial determinants in food allergy. Immunity 53, 277–289 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Abdel-Gadir, A. et al. Microbiota therapy acts via a regulatory T cell MyD88/RORγt pathway to suppress food allergy. Nat. Med. 25, 1164–1174 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Iweala, O. I. & Nagler, C. R. The microbiome and food allergy. Annu. Rev. Immunol. 37, 377–403 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hwang, D. W., Nagler, C. R. & Ciaccio, C. E. New and emerging concepts and therapies for the treatment of food allergy. Immunother. Adv. 2, ltac006 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  10. 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).

    Article  CAS  PubMed  Google Scholar 

  11. Vasapolli, R. et al. Analysis of transcriptionally active bacteria throughout the gastrointestinal tract of healthy individuals. Gastroenterology 157, 1081–1092. e1083 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Lkhagva, E. et al. The regional diversity of gut microbiome along the GI tract of male C57BL/6 mice. BMC Microbiol. 21, 44 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yuan, C., Graham, M., Staley, C. & Subramanian, S. Mucosal microbiota and metabolome along the intestinal tract reveal a location-specific relationship. mSystems 5, e00055-20 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yu, W., Freeland, D. M. H. & Nadeau, K. C. Food allergy: immune mechanisms, diagnosis and immunotherapy. Nat. Rev. Immunol. 16, 751–765 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Feehley, T. et al. Healthy infants harbor intestinal bacteria that protect against food allergy. Nat. Med. 25, 448–453 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Shan, M. et al. Mucus enhances gut homeostasis and oral tolerance by delivering immunoregulatory signals. Science 342, 447–453 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Woting, A. & Blaut, M. Small intestinal permeability and gut-transit time determined with low and high molecular weight fluorescein isothiocyanate-dextrans in C3H mice. Nutrients 10, 685 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Caride, V. et al. Scintigraphic determination of small intestinal transit time: comparison with the hydrogen breath technique. Gastroenterology 86, 714–720 (1984).

    Article  CAS  PubMed  Google Scholar 

  19. Chehade, M. & Mayer, L. Oral tolerance and its relation to food hypersensitivities. J. Allergy Clin. Immunol. 115, 3–12 (2005).

    Article  PubMed  Google Scholar 

  20. Han, K. et al. Generation of systemic antitumour immunity via the in situ modulation of the gut microbiome by an orally administered inulin gel. Nat. Biomed. Eng. 5, 1377–1388 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Osterfeld, H. et al. Differential roles for the IL-9/IL-9 receptor α-chain pathway in systemic and oral antigen–induced anaphylaxis. J. Allergy Clin. Immunol. 125, 469–476.e462 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ahrens, R. et al. Intestinal mast cell levels control severity of oral antigen-induced anaphylaxis in mice. Am. J. Pathol. 180, 1535–1546 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Jones, S. M. & Burks, A. W. Food allergy. N. Engl. J. Med. 377, 1168–1176 (2017).

    Article  PubMed  Google Scholar 

  24. Galand, C. et al. IL-33 promotes food anaphylaxis in epicutaneously sensitized mice by targeting mast cells. J. Allergy Clin. Immunol. 138, 1356–1366 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wagenaar, L. et al. Dietary supplementation with nondigestible oligosaccharides reduces allergic symptoms and supports low dose oral immunotherapy in a peanut allergy mouse model. Mol. Nutr. Food Res. 62, 1800369 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Mazzini, E., Massimiliano, L., Penna, G. & Rescigno, M. Oral tolerance can be established via gap junction transfer of fed antigens from CX3CR1+ macrophages to CD103+ dendritic cells. Immunity 40, 248–261 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Wang, S. H. et al. An exhausted phenotype of TH2 cells is primed by allergen exposure, but not reinforced by allergen‐specific immunotherapy. Allergy 76, 2827–2839 (2021).

    Article  CAS  PubMed  Google Scholar 

  28. Huang, C.-H., Wang, C.-C., Lin, Y.-C., Hori, M. & Jan, T.-R. Oral administration with diosgenin enhances the induction of intestinal T helper 1-like regulatory T cells in a murine model of food allergy. Int. Immunopharmacol. 42, 59–66 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Hong, J. Y. et al. Frontline science: TLR3 activation inhibits food allergy in mice by inducing IFN‐γ+ Foxp3+ regulatory T cells. J. Leukoc. Biol. 106, 1201–1209 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Burks, A. W. et al. Oral immunotherapy for treatment of egg allergy in children. N. Engl. J. Med. 367, 233–243 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kastl, A. J. Jr, Terry, N. A., Wu, G. D. & Albenberg, L. G. The structure and function of the human small intestinal microbiota: current understanding and future directions. Cell. Mol. Gastroenterol. Hepatol. 9, 33–45 (2020).

    Article  PubMed  Google Scholar 

  32. El Aidy, S., Van Den Bogert, B. & Kleerebezem, M. The small intestine microbiota, nutritional modulation and relevance for health. Curr. Opin. Biotechnol. 32, 14–20 (2015).

    Article  PubMed  Google Scholar 

  33. Wang, K. et al. Gut microbiota disorder caused by diterpenoids extracted from Euphorbia pekinensis aggravates intestinal mucosal damage. Pharmacol. Res. Perspect. 9, e00765 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ma, L. et al. Xuanfei Baidu decoction attenuates intestinal disorders by modulating NF-κB pathway, regulating T cell immunity and improving intestinal flora. Phytomedicine 101, 154100 (2022).

    Article  CAS  PubMed  Google Scholar 

  35. Ling, Z. et al. Altered fecal microbiota composition associated with food allergy in infants. Appl. Environ. Microbiol. 80, 2546–2554 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Matysiak-Budnik, T. et al. Gastric Helicobacter infection inhibits development of oral tolerance to food antigens in mice. Infect. Immun. 71, 5219–5224 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chen, M. et al. Nasal bacterial microbiome differs between healthy controls and those with asthma and allergic rhinitis. Front. Cell. Infect. Microbiol. 12, 841995 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Riskumäki, M. et al. Interplay between skin microbiota and immunity in atopic individuals. Allergy: Eur. J. Allergy. Clin. Immunol. 76, 1280–1284 (2021).

    Article  Google Scholar 

  39. Achreja, A. et al. Metabolic collateral lethal target identification reveals MTHFD2 paralogue dependency in ovarian cancer. Nat. Metab. 4, 1119–1137 (2022).

    Article  CAS  PubMed  Google Scholar 

  40. Zhu, Z. et al. Tumour-reprogrammed stromal BCAT1 fuels branched-chain ketoacid dependency in stromal-rich PDAC tumours. Nat. Metab. 2, 775–792 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bettio, L. E., Gil-Mohapel, J. & Rodrigues, A. L. S. Guanosine and its role in neuropathologies. Purinergic Signal. 12, 411–426 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Luo, Y. et al. Guanosine and uridine alleviate airway inflammation via inhibition of the MAPK and NF-κB signals in OVA-induced asthmatic mice. Pulm. Pharmacol. Ther. 69, 102049 (2021).

    Article  CAS  PubMed  Google Scholar 

  43. The Integrative HMP (iHMP) Research Network Consortium. The integrative human microbiome project. Nature 569, 641–648 (2019).

  44. Rahnavard, G. et al. High-sensitivity pattern discovery in large multi’omic datasets. https://huttenhower.sph.harvard.edu/halla (2017).

  45. Zhang, H., Hui, X., Wang, Y. & Lu, X. Angong Niuhuang Pill ameliorates cerebral ischemia/reperfusion injury in mice partly by restoring gut microbiota dysbiosis. Front. Pharmacol. 13, 1001422 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Morafo, V. et al. Genetic susceptibility to food allergy is linked to differential TH2-TH1 responses in C3H/HeJ and BALB/c mice. J. Allergy Clin. Immunol. 111, 1122–1128 (2003).

    Article  CAS  PubMed  Google Scholar 

  47. Wagenaar, L. et al. Mouse strain differences in response to oral immunotherapy for peanut allergy. Immun. Inflamm. Dis. 7, 41–51 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Arifuzzaman, M. et al. Inulin fibre promotes microbiota-derived bile acids and type 2 inflammation. Nature 611, 578–584 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Barletta, B. et al. Probiotic VSL# 3‐induced TGF‐β ameliorates food allergy inflammation in a mouse model of peanut sensitization through the induction of regulatory T cells in the gut mucosa. Mol. Nutr. Food Res. 57, 2233–2244 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Gong, W. et al. Cancer-specific type-I interferon receptor signaling promotes cancer stemness and effector CD8+ T-cell exhaustion. Oncoimmunology 10, 1997385 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Kozich, J. J., Westcott, S. L., Baxter, N. T., Highlander, S. K. & Schloss, P. D. Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl. Environ. Microbiol. 79, 5112–5120 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Percy, A. J., Proos, R., Demianova, Z., Backiel, K. & Ubhi, B. K. Standardizing Quantitative Metabolomics Analyses Through the QReSS™ Kit.

  53. Marvar, J. et al. Porous PDMS‐based microsystem (ExoSponge) for rapid cost‐effective tumor extracellular vesicle isolation and mass spectrometry‐based metabolic biomarker screening. Adv. Mater. Technol. 8, 2201937 (2023).

    Article  CAS  Google Scholar 

  54. Pang, Z. et al. Using MetaboAnalyst 5.0 for LC–HRMS spectra processing, multi-omics integration and covariate adjustment of global metabolomics data. Nat. Protoc. 17, 1735–1761 (2022).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Institutes of Health (NIH) (R01DK125087, R01DE030691, R01DE026728, R01DE031951, R01NS122536, R01CA210273, R01CA271799, R01CA271369, R01CA227622, R01CA222251, R01CA204969 and P30CA046592 to J.J.M. and U01DE033330 to Y.L.L.). Y. Xie is supported by NIH funding R01HL166508 and an NSF award IOS 2107215. D.N. is supported by a Rogel Cancer Center grant and a Forbes Scholar Award. We thank the NIH Tetramer Core Facility (contract HHSN272201300006C) for provision of MHC-I tetramers.

Author information

Authors and Affiliations

Authors

Contributions

K.H., F.X. and J.J.M. designed the study. K.H. and F.X. performed the experiments. O.A., M.N., F.W., K.O., A.A., S.C. and D.N. assisted with the measurement and analysis of short-chain fatty acids and untargeted metabolomics. O.A. performed the machine learning analysis. J.J.O. assisted with the casein and peanut allergy model studies and provided useful discussion. S.K., Y.K. and N.K. assisted with the small intestine studies. M.T.P., Y.S.C., H.D. and Q.W. provided technical help with immune cell analyses in the colon and lungs. J.X., X.Z., X.H., J.A., M.T.P., X.A., Y. Xu and S.-H.L. provided technical help with flow cytometry, NMR and MALDI-TOF. A.K. assisted in the rheology test. Z.L., W.G., Y. Xie and Y.L.L. assisted with the scRNA-seq experiments and analysis. K.H., F.X. and J.J.M. interpreted the data and wrote the paper.

Corresponding author

Correspondence to James J. Moon.

Ethics declarations

Competing interests

Patent applications for inulin-gel-based treatment for allergies and other disorders have been filed in the USA, Europe, Japan, South Korea, China, Australia and Canada (US20230346826, WO2022066825, US20220347294 and WO2021061789), with J.J.M., K.H., F.X., J.X., X.H. and X.Z. as inventors. J.J.M. declares financial interests for board membership, as a paid consultant, for research funding, and/or as an equity holder in EVOQ Therapeutics and Saros Therapeutics. The University of Michigan has a financial interest in EVOQ Therapeutics, Inc. The other authors declare no competing interests.

Peer review

Peer review information

Nature Materials thanks Nicola Harris, Tian Xia and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Biosafety evaluation.

BALB/c mice were treated as in Fig. 1b. The bodyweight changes during OIT (a) and body weight on day 48 (b) were recorded. (c) Blood samples were collected for CBC test on day 48. (d) Serum samples were collected for biochemistry test on day 48. Data represent mean ± s.e.m. (n = 5 for Naive, 7 for Inulin gel/OVA (a), n = 5 for Naive, 8 for all other groups (b,c), n = 5 for Naive, 8 for all other groups in ALT and AST datasets, 3 for Naive, 8 for PBS or 7 for all other groups in BUN dataset, 2 for Naive, 8 for PBS and OVA, or 6 for Inulin gel/OVA in CREA dataset (d) biologically independent samples). ns: not significant. Data were analysed by one-way (b-d) or two-way ANOVA (a) with Bonferroni’s multiple comparisons test.

Extended Data Fig. 2 Inulin gel/OVA OIT protects against OVA challenges.

a, Therapeutic regimen. BALB/c mice were sensitized with alum/OVA on days 0 and 14. From day 29, the mice were orally gavaged with inulin gel (55 mg per dose), or inulin gel (55 mg per dose)/OVA (1 mg per dose). From day 49, mice were intragastrically (i.g.) challenged with OVA (50 mg per dose) on 6 alternating days. Shown are the (b) average body temperature change after the 6th challenge, (c) anaphylactic scores, (d) diarrhea occurrence rate, and (e) diarrhea severity during challenges. f, Diarrhea-induced body weight change after the 6th i.g. challenge. g, OVA-specific IgE levels in serum on days 48 and 55. Data represent mean ± s.e.m. (n = 8 biologically independent samples). Data were analysed by two-way ANOVA (b,c,e) with Bonferroni’s multiple comparisons test, or unpaired, two-sided Student’s t-test (f,g).

Extended Data Fig. 3 DP values of inulin affect OIT efficacy.

a, The DP values of inulin from different vendors were measured by 1HNMR and MALDI-TOF. b, BALB/c mice were treated as in Fig. 1b and received OIT treatments of inulin (DP7)/OVA, inulin gel (DP10)/OVA, and inulin gel (DP23)/OVA, respectively. Mice were i.g. challenged for 6 times. The body temperatures during the 6th i.g. challenge, the anaphylactic symptoms, diarrhea occurrence rate and severity during the challenges were recorded. c, The diarrhea images at the 3rd i.g. challenge. d, Average mast cells and the representative images in ileum. Data represent mean ± s.e.m. (In body temperature, n = 7 for Inulin (DP7)/OVA and Inulin gel (DP10)/OVA, or 8 for Inulin gel (DP23)/OVA, in all other datasets, n = 8 (b), n = 6 for Inulin (DP7)/OVA, Inulin gel (DP10)/OVA, 4 for Inulin gel (DP23)/OVA (d) biologically independent samples). ns, not significant. Data were analysed using two-way ANOVA (b), or one-way ANOVA (d) with Bonferroni’s multiple comparisons test.

Extended Data Fig. 4 In vitro Treg cell differentiation.

BMDCs were treated with inulin gel/OVA or inulin gel/OVA-II peptide for 24 h. BMDCs were washed and co-cultured with CFSE-labeled OT-II CD4+ T cells. Shown are the frequency of Foxp3+CD4+ T cells, proliferating CFSElow cells, and representative flow cytometry plots after 5 days of co-culture. Data represent mean ± s.e.m. (n = 4 biologically independent samples). Data were analysed by one-way ANOVA with Bonferroni’s multiple comparisons test.

Extended Data Fig. 5 Long-term protection.

Alum/OVA-sensitized mice received the indicated OIT treatment, followed by 4 times of i.g. challenges of OVA protein. The doses of inulin and OVA were 55 mg and 1 mg, respectively. All treatments were discontinued from day 55. From day 146, the mice were i.g. challenged with OVA. Shown are the body temperature drop, anaphylactic score, diarrhea score, and body weight change during the 5th i.g. re-challenge. Data represent mean ± s.e.m. (n = 5 for Naive, 7 for Inulin/OVA or 6 for all other groups, biologically independent samples). Data were analysed by one-way or two-way ANOVA with Bonferroni’s multiple comparisons test.

Extended Data Fig. 6 Microbes abundances in ileal contents.

Alum/OVA-sensitized BALB/c mice were treated as in Fig. 1b. Shown are the relative abundances of several genera in the ileal contents. Data represent mean ± s.e.m. (n = 5 for Naive, 7 for OVA or 8 for all other groups, biologically independent samples). Data were analysed by one-way ANOVA with Bonferroni’s multiple comparisons test.

Extended Data Fig. 7 Immunomodulatory guanosine as identified by metabolomics and machine learning based analyses.

Alum/OVA-sensitized BALB/c mice were treated as in Fig. 1b. a-b, Untargeted metabolome analysis in the ileal contents and feces. Shown are the sPL-SDA analysis of the metabolites in the ileal contents on day 49 and feces on day 48 (a), fold changes of metabolites (whose P-value is less than 0.05) in the ileal and fecal contents. The size of the circle denotes the fold change of upregulated metabolites in the inulin gel/OVA group, compared with naïve, PBS, OVA, and inulin/OVA groups. The P-values are unadjusted and are based on a two-sided t-test statistics (b). c-e, Metabolite-induced differentiation of CD4+ T cells in vitro. CD4+ T cells isolated from Foxp3(GFP) reporter mice were cultured with various metabolites in suboptimal Treg-inducing condition (1 µg/mL anti-CD3 antibody, 1 µg/mL anti-CD28 antibody, 5 ng/mL IL-2, and 1 ng/mL TGF-β) and analyzed on day 3 for Foxp3 expression (c). CD4+ T cells isolated from wide-type (WT) C57BL/6 mice were cultured with various metabolites in TH1-inducing condition (1 µg/mL anti-CD3 antibody, 1 µg/mL anti-CD28 antibody, 5 ng/mL IL-2, 10 ng/mL IL-12p40, and 10 μg/mL anti-IL-4 antibody) and analyzed on day 5 for IFN-γ secretion (d). CD4+ T cells isolated from WT C57BL/6 mice were cultured with various metabolites in TH2-inducing condition (1 µg/mL anti-CD3 antibody, 1 µg/mL anti-CD28 antibody, 5 ng/mL IL-2, 10 ng/mL IL-4, and 10 μg/mL anti-IFN-γ antibody) and analyzed on day 5 for IFN-γ secretion (e). f, Machine learning-based association study between ileal metabolomics and microbes at the family level. Only features with significant clusters were shown on the heatmap (Hallagram). g, Spearman’s correlation coefficient analyses between the relative abundances of ileal guanosine and Eggerthellaceae (family) or Enterorhabdus (genus). Data represent the mean ± s.e.m. from one of two independent experiments (n = 5 for Naive or 8 for all other groups (a), n = 6 for PBS and DMSO, or 4 for all other groups (c), n = 6 for PBS and DMSO, or 3 for all other groups (d,e) biologically independent samples). Data were analysed by one-way ANOVA with Bonferroni’s multiple comparisons test (c-e), or two-tailed Spearman’s rank correlation test (g).

Extended Data Fig. 8 OVA/guanosine OIT protects mice against allergen challenges.

a, Schema of the intestinal anaphylaxis and therapeutic regimen of OVA/guanosine OIT. BALB/c mice were sensitized with alum/OVA on days 0 and 14. From day 28, mice were orally gavaged with OVA (1 mg per dose) and provided with normal drinking water either with or without 37.5 µg/mL of guanosine (denoted OVA and OVA/guanosine, respectively). Inulin gel (55 mg per dose)/OVA (1 mg per dose) treated mice that were provided with normal drinking water were included as an additional control. From day 48, mice were i.g. challenged with OVA (50 mg per dose) on 4 alternating days. b, After the 4th i.g. challenge, changes in core body temperature were measured. c-e, Over the 4 consecutive i.g. challenges, animals were analyzed for anaphylactic scores (c), diarrhea severity score (d), and body weight change (e). Data represent the mean ± s.e.m. (n = 8 for Inulin gel/OVA or 7 for all other groups (b), n = 8 (c-e) biologically independent samples). Data were analysed by two-way ANOVA with Bonferroni’s multiple comparisons test.

Extended Data Fig. 9 Less frequent inulin gel/OVA OIT remains protective.

Schema of the intestinal anaphylaxis and therapeutic regimen. BALB/c mice were sensitized with alum/OVA on days 0 and 14. From day 29, mice were orally gavaged with PBS, OVA (1 mg per dose), or inulin gel (55 mg per dose)/OVA (1 mg per dose) every other day. From day 40, mice were i.g. challenged with OVA (50 mg per dose) on 6 alternating days. b, After the 6th i.g. challenge, changes in the average core body temperature were measured. c-f, Over the 6 consecutive i.g. challenges, animals were analyzed for anaphylactic scores (c), diarrhea severity score (d), diarrhea occurrence rate (e), and bodyweight drop (f). Data represent mean ± s.e.m. (n = 5 for Naive, 8 for PBS or 9 for Inulin gel/OVA (alternative) (b-f) biologically independent samples). Data were analysed by two-way ANOVA (b-d,f) with Bonferroni’s multiple comparisons test.

Extended Data Fig. 10 Inulin gel does not induce type 2 inflammatory response.

Naive C57BL/6 mice were fed with high (26%) inulin chow diet, control chow diet (0% inulin, PBS group), control chow diet plus inulin (55 mg/dose, 3 times per 4 days, inulin group), or control chow diet plus inulin gel (55 mg/dose, 3 times per 4 days, inulin gel group). On day 13, the frequencies of immune cells in the colon were detected: (a) eosinophils; (b) neutrophils; (c) Ly6Chigh monocytes; (d) CD64+ macrophages; (e) DCs; (f) B cells; (g) T cells. (h) The frequency of eosinophils in the lung. (i) The representative flow plot of eosinophils in the colon and lung. The concentrations of ω-MCA, CA, TCA, UDCA, DCA, and CDCA in the (j) serum, (k) feces, and (l) cecum on day 13. Data represent mean ± s.e.m. (n = 8 biologically independent samples). ns: not significant. Data were analysed by (a-h) one-way, or (j-l) two-way ANOVA with Bonferroni’s multiple comparisons test.

Supplementary information

Supplementary Information

Supplementary Figs. 1–15, Table 1 and Methods.

Reporting Summary

Source data

Source Data Fig. 1

Raw data points in Excel.

Source Data Fig. 2

Raw data points in Excel.

Source Data Fig. 3

Raw data points in Excel.

Source Data Fig. 4

Raw data points in Excel.

Source Data Fig. 5

Raw data points in Excel.

Source Data Fig. 6

Raw data points in Excel.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Han, K., Xie, F., Animasahun, O. et al. Inulin-gel-based oral immunotherapy remodels the small intestinal microbiome and suppresses food allergy. Nat. Mater. (2024). https://doi.org/10.1038/s41563-024-01909-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41563-024-01909-w

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