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:

Thymic development of gut-microbiota-specific T cells

Abstract

Humans and their microbiota have coevolved a mutually beneficial relationship in which the human host provides a hospitable environment for the microorganisms and the microbiota provides many advantages for the host, including nutritional benefits and protection from pathogen infection1. Maintaining this relationship requires a careful immune balance to contain commensal microorganisms within the lumen while limiting inflammatory anti-commensal responses1,2. Antigen-specific recognition of intestinal microorganisms by T cells has previously been described3,4. Although the local environment shapes the differentiation of effector cells3,4,5 it is unclear how microbiota-specific T cells are educated in the thymus. Here we show that intestinal colonization in early life leads to the trafficking of microbial antigens from the intestine to the thymus by intestinal dendritic cells, which then induce the expansion of microbiota-specific T cells. Once in the periphery, microbiota-specific T cells have pathogenic potential or can protect against related pathogens. In this way, the developing microbiota shapes and expands the thymic and peripheral T cell repertoire, allowing for enhanced recognition of intestinal microorganisms and pathogens.

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: Commensal colonization leads to an expansion of bacteria-specific T cells in the thymus.
Fig. 2: Enrichment of CX3CR1+ DCs in the thymus after colonization with commensal microorganisms.
Fig. 3: Thymic microbiota-specific T cells are functional in distal tissues.

Similar content being viewed by others

Data availability

All data generated and supporting the findings of this study are available within the paper. The RNA-seq data have been deposited in the Gene Expression Omnibus (GEO) under accession number GSE171279. The 16S sequencing data have been submitted to the Sequence Read Archive (SRA) under the BioProject ID 718898. Additional information and materials will be made available upon request. Source data are provided with this paper.

References

  1. Hooper, L. V., Littman, D. R. & Macpherson, A. J. Interactions between the microbiota and the immune system. Science 336, 1268–1273 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Zegarra-Ruiz, D. F. et al. A diet-sensitive commensal Lactobacillus strain mediates TLR7-dependent systemic autoimmunity. Cell Host Microbe 25, 113–127 (2019).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Yang, Y. et al. Focused specificity of intestinal TH17 cells towards commensal bacterial antigens. Nature 510, 152–156 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bäckhed, F. et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe 17, 690–703 (2015).

    Article  PubMed  Google Scholar 

  7. Boehm, T. & Swann, J. B. Thymus involution and regeneration: two sides of the same coin? Nat. Rev. Immunol. 13, 831–838 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Cebula, A. et al. Thymus-derived regulatory T cells contribute to tolerance to commensal microbiota. Nature 497, 258–262 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kotov, D. I. & Jenkins, M. K. Peptide:MHCII tetramer-based cell enrichment for the study of epitope-specific CD4+ T cells. Curr. Protoc. Immunol. 125, e75 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Sakaguchi, S. Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat. Immunol. 6, 345–352 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Egawa, T. & Littman, D. R. ThPOK acts late in specification of the helper T cell lineage and suppresses Runx-mediated commitment to the cytotoxic T cell lineage. Nat. Immunol. 9, 1131–1139 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hogquist, K. A., Xing, Y., Hsu, F.-C. & Shapiro, V. S. T cell adolescence: maturation events beyond positive selection. J. Immunol. 195, 1351–1357 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Anderson, K. G. et al. Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protocols 9, 209–222 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Owen, D. L. et al. Thymic regulatory T cells arise via two distinct developmental programs. Nat. Immunol. 20, 195–205 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chopp, L. B. et al. An integrated epigenomic and transcriptomic map of mouse and human αβ T cell development. Immunity 53, 1182–1201 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Diehl, G. E. et al. Microbiota restricts trafficking of bacteria to mesenteric lymph nodes by CX3CR1hi cells. Nature 494, 116–120 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Varol, C., Zigmond, E. & Jung, S. Securing the immune tightrope: mononuclear phagocytes in the intestinal lamina propria. Nat. Rev. Immunol. 10, 415–426 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Longman, R. S. et al. CX3CR1+ mononuclear phagocytes support colitis-associated innate lymphoid cell production of IL-22. J. Exp. Med. 211, 1571–1583 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kim, M. et al. Critical role for the microbiota in CX3CR1+ intestinal mononuclear phagocyte regulation of intestinal T cell responses. Immunity 49, 151–163 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hadeiba, H. & Butcher, E. C. Thymus-homing dendritic cells in central tolerance. Eur. J. Immunol. 43, 1425–1429 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Nowotschin, S. & Hadjantonakis, A.-K. Use of KikGR a photoconvertible green-to-red fluorescent protein for cell labeling and lineage analysis in ES cells and mouse embryos. BMC Dev. Biol. 9, 49 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Sohy, D. et al. Hetero-oligomerization of CCR2, CCR5, and CXCR4 and the protean effects of “selective” antagonists. J. Biol. Chem. 284, 31270–31279 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Coombes, J. L., Robinson, N. J., Maloy, K. J., Uhlig, H. H. & Powrie, F. Regulatory T cells and intestinal homeostasis. Immunol. Rev. 204, 184–194 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Di Padova, F. E. et al. A broadly cross-protective monoclonal antibody binding to Escherichia coli and Salmonella lipopolysaccharides. Infect. Immun. 61, 3863–3872 (1993).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Bensinger, S. J., Bandeira, A., Jordan, M. S., Caton, A. J. & Laufer, T. M. Major histocompatibility complex class II-positive cortical epithelium mediates the selection of CD4+25+ immunoregulatory T cells. J. Exp. Med. 194, 427–438 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Klein, L., Kyewski, B., Allen, P. M. & Hogquist, K. A. Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see). Nat. Rev. Immunol. 14, 377–391 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Leech, J. M. et al. Toxin-triggered interleukin-1 receptor signaling enables early-life discrimination of pathogenic versus commensal skin bacteria. Cell Host Microbe 26, 795–809 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Scharschmidt, T. C. et al. A wave of regulatory T cells into neonatal skin mediates tolerance to commensal microbes. Immunity 43, 1011–1021 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hashimoto, K., Joshi, S. K. & Koni, P. A. A conditional null allele of the major histocompatibility IA-beta chain gene. Genesis 32, 152–153 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Jung, S. et al. Analysis of fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol. Cell. Biol. 20, 4106–4114 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Parkhurst, C. N. et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sano, T. et al. An IL-23R/IL-22 circuit regulates epithelial serum amyloid A to promote local effector Th17 responses. Cell 163, 381–393 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Buchan, A., Hadden, M. & Suzuki, M. T. Development and application of quantitative-PCR tools for subgroups of the Roseobacter clade. Appl. Environ. Microbiol. 75, 7542–7547 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chern, E. C., Siefring, S., Paar, J., Doolittle, M. & Haugland, R. A. Comparison of quantitative PCR assays for Escherichia coli targeting ribosomal RNA and single copy genes. Lett. Appl. Microbiol. 52, 298–306 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Valdez, Y. et al. Nramp1 expression by dendritic cells modulates inflammatory responses during Salmonella Typhimurium infection. Cell. Microbiol. 10, 1646–1661 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Moon, J. J. et al. Tracking epitope-specific T cells. Nat. Protocols 4, 565–581 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. Moon, J. J. et al. Naive CD4+ T cell frequency varies for different epitopes and predicts repertoire diversity and response magnitude. Immunity 27, 203–213 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chu, H. H., Moon, J. J., Kruse, A. C., Pepper, M. & Jenkins, M. K. Negative selection and peptide chemistry determine the size of naive foreign peptide-MHC class II-specific CD4+ T cell populations. J. Immunol. 185, 4705–4713 (2010).

    Article  CAS  PubMed  Google Scholar 

  39. Viladomiu, M. et al. IgA-coated E. coli enriched in Crohn’s disease spondyloarthritis promote TH17-dependent inflammation. Sci. Transl. Med. 9, eaaf9655 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Engström, P. G. et al. Systematic evaluation of spliced alignment programs for RNA-seq data. Nat. Methods 10, 1185–1191 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 35, 166–169 (2015).

    Article  Google Scholar 

  43. Huang, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protocols 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  44. Berg, D. J. et al. Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4+ TH1-like responses. J. Clin. Invest. 98, 1010–1020 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grants R01AI136963 (G.E.D. and M.L.B.), R01AI125264 (G.E.D.), R01DK114456 (M.L.B.), R01AI130152 (T.E.) and R01 DK114252 (R.S.L.); the Kleberg Foundation (G.E.D. and M.L.B); the Kenneth Rainin Foundation (G.E.D.); the Leukemia and Lymphoma Society Scholar Award (T.E.); and the Cytometry and Cell Sorting Core at Baylor College of Medicine, with funding from the CPRIT Core Facility Support Award (CPRIT-RP180672) and the NIH (P30 CA125123 and S10 RR024574) and with the assistance of J. M. Sederstrom. We thank the Baylor College of Medicine Genetically Engineered Mouse Core supported by the Cancer Center Grant (P30 CA125123); the Mouse Embryonic Stem Cell Core at Baylor College of Medicine; D. Littman and C.-S. Hsieh for critical reading of the manuscript; N. Ajami for advice on 16S; and the NIH Tetramer Facility, which is supported by contract HHSN272201300006C from the NIAID.

Author information

Authors and Affiliations

Authors

Contributions

D.F.Z.-R., T.E., R.S.L., M.L.B. and G.E.D. designed the experiments and analysed data. D.F.Z.-R. and G.E.D. wrote the manuscript with input from all co-authors. D.F.Z.-R., D.V.K., K.N., M.K, W.-J.H.W., F.B.S.-M., A.A.H., S.M., S.O., M.L.B. and G.E.D. conducted experiments and analysed data. R.B. and J.L.R. were responsible for germ-free mouse experiments.

Corresponding authors

Correspondence to Matthew L. Bettini or Gretchen E. Diehl.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Dennis Kasper and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Characterization of SFB-tetramer+ CD4+ T cells.

a, Gating strategy used to identify SFB-tetramer+ CD4+ T cells after magnetic enrichment. Live, CD45+ single cells negative for CX3CR1, Ly6G, B220, Ly6C, MHCII and CD8 were further analysed. From the CD3+CD4+ population, SFB-specific cells were identified as double positive for APC- and PE- (Fig. 1) or single positive for APC- or PE-conjugated tetramers specific for SFB peptide QFSGAVPNKTD (all other figures). b, Mice were colonized with SFB at weaning and two weeks later thymic T cells were compared to age-matched control (CTRL) mice. Frequencies and counts of SFB-tetramer+ cells in thymic flow-through after magnetic enrichment (n = 3 per group). c, 7B8 TCR transgenic mice were left uncolonized or SFB-colonized at weaning and two weeks later CD4+ counts in the thymus were compared (control n = 6; SFB n = 7). df, Mice were colonized with SFB at weaning and two weeks later thymic T cells were compared to age-matched control mice. d, Frequencies and counts of SFB-tetramer+ cells in CD8+ (single-positive; SP) and CD4+CD8+ (double-positive; DP) T cells (n = 10 per group). e, Frequencies and counts of no tetramer (NT), 2W1S, H. hepaticus (HH) and human (HU) tetramer+ cells in CD4+ T cells (n = 5 per group). f, Frequencies and counts of CD4+, CD8+ and CD4+CD8+ thymic subsets. gk, Representative flow plots of control and SFB CD4+ T cells for RORγt and FOXP3 (g), CD25 (h), CD44 (i); CD24 and TCRβ (j) and CD69 and TCRβ (k). In fk, n = 15 per group. Each replicate is a biologically independent sample. Data are shown as individual values and mean; P values by two-tailed unpaired t-test (b, c) or two-way ANOVA with Sidak’s post hoc test (df).

Source data

Extended Data Fig. 2 Thymic expansion of bacteria-specific T cells after colonization with commensal microorganisms at weaning.

ac, Thymic SFB-tetramer+ cells from young mice were analysed by flow cytometry (n = 15). a, CD24 and TCRβ. b, CD44. c, CD69 and TCRβ. d, Mice were colonized with SFB at weaning for two weeks and were injected intravenously with anti-CD45 before euthanasia. Flow cytometry of CD45 in thymic SFB-tetramer+ cells and spleen (n = 5). e, RAG2-GFP mice were colonized with SFB at weaning and two weeks later thymi were collected. Flow cytometry of thymic CD4+ cells (n = 7). f, g, Thymic SFB-non-specific and SFB-specific CD4+ T cells from young mice two weeks after colonization were analysed by RNA-seq. f, Principal component analysis of SFB-non-specific and SFB-specific CD4+ T cells (circled) compared to thymocytes at different developmental stages (GSE148973). g, Heat map showing row-standardized z-scores of mRNA expression for genes in enriched KEGG pathways (P < 0.05). In f, g, n = 5 (SFB-non-specific T cells); n = 3 (SFB-specific T cells). Each SFB-specific T cell replicate is a pool of 10 thymi. hj, Mice were colonized with SFB at weaning and were compared four weeks later to age-matched control mice. h, Flow cytometry and counts of SFB-tetramer+ cells in the MLNs (n = 5 (CTRL; n = 15 (SFB)), spleen (SPL; n = 5 per group), ileum (n = 5 (CTRL); n = 10 (SFB)) and colon (n = 5 per group). i, j, Flow cytometry (i) and counts (j) of thymic SFB-tetramer+ cells (n = 5 (CTRL); n = 15 (SFB)). k, RAG2-GFP mice were colonized with SFB at weaning and four weeks later thymi were collected. Flow cytometry of thymic CD4+ cells (n = 7). l, m, SPF and germ-free mice were treated with antibiotics (ABX) at weaning and compared two weeks later to age-matched control mice. Counts of total thymocytes, CD4+, CD8+ and thymic Treg cells in SPF mice (l; n = 15 (CTRL); n = 9 (ABX)) and germ-free mice (m; n = 3 (CTRL); n = 4 (ABX)). Each replicate is a biologically independent sample. Data are shown as individual values and mean; P values by two-tailed unpaired t-test (j, l, m) or two-way ANOVA with Sidak’s post hoc test (h).

Source data

Extended Data Fig. 3 Bacterial DNA is found in the thymus after early-life colonization.

a, b, hj, Mice were colonized with SFB or E. coli at weaning (young) or at 12 weeks of age (adult) and one week later were compared to age-matched control mice. a, b, SFB-specific qPCR in faecal pellets (FP) (a; young: CTRL, SFB n = 8; adult: CTRL, SFB n = 6); and MLNs (b; young: CTRL, SFB n = 9; adult: CTRL, SFB n = 7). hj, E. coli-specific qPCR in the thymus (h; young: CTRL n = 10, EC n = 15; adult: CTRL, EC n = 15), MLNs (i; young: CTRL, EC n = 8; adult: CTRL, EC n = 7) and faecal pellets (j; young: CTRL, EC n = 8; adult: CTRL, EC n = 10). Mice were colonized with SFB at weaning and compared one week later to age-matched control mice. c, e, SFB-specific qPCR (c; n = 6 per group) and 16S qPCR (e) in the thymus, MLNs, liver, spleen, heart and lungs (in e, thymus, MLNs: CTRL, SFB n = 12; liver, SPL, heart, lungs: CTRL, SFB n = 6). d, g, Mice were colonized with SFB at weaning and analysed up to three weeks later. SFB-specific PCR (d) and 16S qPCR (g) in the thymus, MLNs and faecal pellets (n = 5 per group). f, Mice were colonized with SFB at 12 weeks of age and compared one week later to age-matched control mice. 16S qPCR in thymus and MLNs (thymus: CTRL n = 16, SFB n = 11; MLNs CTRL, SFB n = 7). k, Mice were euthanized at 4 weeks of age, tissues were collected, DNA was isolated and 16S rDNA sequencing was performed. Bacterial phyla and class relative abundance in the thymus, MLNs and caecum (n = 5 per group; thymus and MLNs were pooled). ln, Mice were treated with antibiotics at weaning and compared one week later to age-matched control mice. 16S qPCR in the thymus (l; n = 7 (CTRL), n = 8 (ABX)); MLNs (m; n = 16 (CTRL), n = 9 (ABX)) and faecal pellets (n; n = 10 per group). Each replicate is a biologically independent sample. Data are shown as mean ± s.e.m. (d, g) or as individual values and mean; P values by two-tailed unpaired t-test (ln) or two-way ANOVA with Tukey’s (a, b, f, hj), Sidak’s (c, e) or Fisher’s LSD (e) post hoc test.

Source data

Extended Data Fig. 4 CX3CR1+ DCs are enriched in the thymus after colonization with commensal microorganisms in young mice.

ac, g, Thymic cell populations were analysed at weaning (W3), one (W4) and two (W5) weeks after weaning, and at 12 weeks of age (W12). Frequencies and counts of CD4+, CD8+ and CD4+CD8+ subsets (a); Treg cells (b) (in a, b, W3 n = 5; W4 n = 11; W5 n = 15; W12 n = 8); CD11c+MHCII+ DCs (c); and CD103+ and CX3CR1+ DC subsets (g) (in c, g, n = 10 (W3); n = 15 (W4, W5, W12)). d, m, p, q, Mice were treated with antibiotics at weaning and thymi were compared two weeks later to age-matched control mice. d, m, Frequencies and counts of CD11c+MHCII+ DCs (d) and CD103+ and CX3CR1+ DC subsets (m) (n = 23 per group). p, q, Frequencies of plasmacytoid DCs (pDCs) (p) and B cells (q) (n = 15 per group). e, f, il, n, o, Mice were colonized with SFB or E. coli at weaning (young) or at 12 weeks of age (adult) and thymi were compared two weeks later to age-matched control mice. e, f, Flow cytometry (e) and counts (f) of CD11c+MHCII+ DCs. i, Representative flow plot of CX3CR1+ DCs. j, Frequencies and counts of CD103+ DCs in the thymus (in e, f, i, j, young: CTRL n = 15; SFB n = 20; EC n = 12. Adult: CTRL n = 20; SFB n = 20; EC n = 16). k, l, Frequencies of CD11c+MHCII+ DCs (k) and CX3CR1+ DC subsets (l) in the MLNs (young: CTRL n = 12; SFB n = 15; EC n = 10. Adult: CTRL n = 8; SFB n = 12; EC n = 8). n, o, Frequencies of pDCs (n) and B cells (o) in the thymus (n = 12 per group). h, Mice were colonized with SFB at weaning for two weeks and were injected intravenously with anti-CD45 before euthanasia. Flow cytometry of CD45 in thymic CX3CR1+ DCs and spleen (n = 5). Each replicate is a biologically independent sample. Data are shown as individual values and mean; P values by two-tailed unpaired t-test (d, p, q), one-way ANOVA with Tukey’s post hoc test (b, c) or two-way ANOVA with Tukey’s (a, f, g, jl, n, o) or Sidak’s (m) post hoc test.

Source data

Extended Data Fig. 5 MHCII expression on CX3CR1+ cells is required for the thymic expansion of bacteria-specific T cells.

ag, Wild-type and CX3-DTR mice were colonized with SFB or E. coli at weaning or left untreated and tissues were compared one week (qPCR) or two weeks (flow cytometry analysis) later. a, Flow cytometry of thymic SFB-tetramer+ cells (CTRL: n = 5; SFB: WT n = 6; CX3-DTR n = 8). b, c, 16S qPCR in the thymus (b; n = 8 (WT); n = 9 (CX3-DTR)); and MLNs (c; n = 13 (WT); n = 12 (CX3-DTR)). d, e, SFB-specific qPCR in the thymus (d; n = 5 per group) and faecal pellets (e; n = 10 (WT); n = 5 (CX3-DTR)). f, g, E. coli-specific qPCR in the thymus (f; WT n = 7; CX3-DTR n = 12) and faecal pellets (g; n = 10). h, Diphtheria-toxin-treated littermate wild-type mice and mice depleted of CD103+ DCs (CD103-DTR CD11c-Cre) were analysed two weeks after weaning. Flow cytometry of CD103+ and CX3CR1+ DC subsets in colon lamina propria (n = 5). i, j, Diphtheria-toxin-treated littermate wild-type, CX3-DTR CD11c-Cre and CD103-DTR CD11c-Cre mice were colonized with E. coli at weaning and thymi were compared one week later. i, E. coli-specific qPCR (n = 8 (WT); n = 10 (CX3-DTR); n = 8 (CD103-DTR)). j, 16S qPCR (n = 24 (WT); n = 10 (CX3-DTR); n = 8 (CD103-DTR)). km, 4-OHT-treated MHCIIfl/+ (WT) and MHCIIfl/− (KO) CX3-Cre mice were colonized with SFB at weaning and thymi were compared two weeks later. k, Frequencies and counts of SFB-tetramer+ cells (n = 10 per group). l, SFB-specific qPCR (n = 5 per group). m, Frequencies of CD103+ and CX3CR1+ DC subsets (n = 10 per group). Each replicate is a biologically independent sample. Data are shown as individual values and mean P values by two-tailed unpaired t-test (bg, k, l) or one-way ANOVA with Tukey’s (i, j) or Sidak’s (m) post hoc test.

Source data

Extended Data Fig. 6 Intestinal CX3CR1+ DCs migrate to the thymus.

a, Thymic DCs were analysed at two weeks after weaning for XCR1 and SIRPα. Flow cytometry of total DCs, CX3CR1+ DCs, CD103+ DCs and pDCs (n = 5). bf, Caeca from KikGR33 transgenic mice were exposed to 405-nm-wavelength light or left untreated at 5 (young) or 14 (adult) weeks of age and two days later the thymus, MLNs and spleen were collected. b, Flow cytometry of RFP+MHCII+ cells in KikGR33 mice without light exposure (n = 5). c, Frequencies and counts of RFP+MHCII+ cells in young mice. d, e, Flow cytometry (d) and counts (e) of RFP+MHCII+ cells in adult mice. f, Flow cytometry of RFP+MHCII+ cells subsets in young and adult thymi. In cf, n = 5 (young); n = 9 (adult). gi, Mice were analysed at weaning (young) or at 12 weeks of age (adult) and thymus chemokine gene expression or colon lamina propria DC populations were analysed. g, Relative expression of chemokines in the thymus (n = 5 (young); n = 15 (adult)). h, CCR5+ cell frequencies of CX3CR1+ DCs in the colon (n = 10 per group). Wild-type and CX3CR1GFP/GFP (KO) mice were colonized with SFB at weaning and treated with TAK-779 or anti-CCL2 antibody and thymi were compared one week later. i, SFB-specific qPCR (n = 17 (WT); n = 13 (anti-CCR5 + anti-CCR2); n = 5 (WT anti-CCR2); n = 10 (KO); n = 5 (KO anti-CCR5 + anti-CCR2)). Each replicate is a biologically independent sample. Data are shown as individual values and mean; P values by two-tailed unpaired t-test (g, h) or one-way ANOVA with Fisher’s LSD post hoc test (i).

Source data

Extended Data Fig. 7 Bacteria-specific T cells from the thymus of young mice are functional in distal tissues.

Wild-type donor mice were colonized with SFB or E. coli at weaning and two weeks later CD4+ T cells from the thymus were sorted and transferred to SFB- or E. coli-colonized Rag2−/− recipient mice. Recipient mice were followed for four weeks and disease severity was compared to Rag2−/− mice receiving age-matched control donor thymocytes. af, SFB (n = 8 (CTRL); n = 13 (SFB)). a, Colon length. b, Immune infiltration in colon lamina propria. c, d, Flow cytometry (c) and counts (d) of TH17 cells. e, Representative flow plot of SFB-tetramer+ cells. f, Flow cytometry of RORγt and T-bet expression in SFB-tetramer+ cells. gi, E. coli (n = 8 (CTRL); n = 15 (EC)). g, Colon length. h, Immune infiltration in colon lamina propria. i, Representative flow plot of TH1 cells. Each replicate is a biologically independent sample. Data are shown as individual values and mean; P values by two-tailed unpaired t-test (a, b, d, g, h).

Source data

Extended Data Fig. 8 Bacteria-specific T cells from the thymus of adult mice do not increase pathology in distal tissues.

Wild-type donor mice were colonized with SFB or E. coli at 12 weeks of age and two weeks later CD4+ T cells from the thymus were sorted and transferred to SFB- or E. coli-colonized Rag2−/− recipient mice. Recipient mice were followed for four weeks and disease severity was compared to Rag2−/− mice receiving age-matched control donor thymocytes. ah, SFB. a, Relative weight change after transfer (n = 5 (CTRL); n = 7 (SFB)). b, Colon length. c, d, Representative H&E (c) and colitis score (d). e, Immune infiltration in colon lamina propria. f, g, Frequencies and counts of TH17 cells (f) and SFB-tetramer+ cells (g). h, Flow cytometry of RORγt and T-bet expression in SFB-tetramer+ cells. In bh, n = 4 (CTRL); n = 7 (SFB). in, E. coli (n = 7 (CTRL); n = 9 (EC)). i, Relative weight change after transfer. j, Colon length. k, l, Representative H&E (k) and colitis score (l). m, Immune infiltration in colon lamina propria. n, Frequencies and counts of TH1 cells. Each replicate is a biologically independent sample. Data are shown as mean ± s.e.m., with P values by two-way ANOVA with Fisher’s LSD post hoc test (a, i), or are shown as individual values and mean, with P values by two-tailed unpaired t-test (b, dg, j, ln). Scale bars, 100 μm (c, k).

Source data

Extended Data Fig. 9 Requirement for antigen presentation by CX3CR1+ cells for thymic T cell pathogenicity in distal tissues.

ad, Diphtheria-toxin-treated littermate SPF wild-type and CX3-DTR donor mice were colonized with SFB at weaning and two weeks later CD4+ T cells from the thymus were sorted and transferred to SFB-colonized Rag2−/− recipient mice. Recipient mice were followed for four weeks and disease severity was assessed. a, Colon length (n = 12 (WT); n = 13 (CX3-DTR). b, Immune infiltration in colon lamina propria. c, d, Flow cytometry (c) and counts (d) of SFB-tetramer+ cells. In bd, n = 8 per group. ek, 4-OHT-treated MHCIIfl/+ (WT) and MHCIIfl/− (KO) CX3-Cre mice were colonized with SFB at weaning and two weeks later CD4+ T cells from the thymus were sorted and transferred to SFB-colonized Rag2−/− recipient mice. Recipient mice were followed for four weeks and disease severity was assessed. e, Relative weight change after transfer. f, Colon length. g, h. Representative H&E (g) and colitis score (h). i, Immune infiltration in colon lamina propria. j, k, Flow cytometry (j) and counts (k) of SFB-tetramer+ cells. In ek, n = 8 (WT); n = 10 (KO). lq, SPF wild-type donor mice were treated with antibiotics at weaning and two weeks later CD4+ T cells from the thymus were sorted and transferred to untreated Rag2−/− recipient mice. Recipient mice were followed for eight weeks and disease severity was compared to Rag2−/− mice receiving age-matched control donor thymocytes. l, Relative weight change after transfer. m, Colon length. n, o, Representative H&E (n) and colitis score (o). p, Immune infiltration in colon lamina propria. q, Frequencies and counts of TH1 cells. In lq, n = 7 (CTRL); n = 9 (ABX). r, Wild-type mice were infected at 5 weeks of age with S. Typhimurium. MLNs were collected and cells restimulated with S. Typhimurium or E. coli. IFN-γ levels in culture supernatants (n = 7 (ST); n = 5 (EC)). Each replicate is a biologically independent sample. Data are shown as mean ± s.e.m., with P values by two-way ANOVA with Fisher’s LSD post hoc test (e, l), or are shown as individual values and mean, with P values by two-tailed unpaired t-test (a, b, d, f, h, i, k, m, or). Scale bars, 100 μm (g, n).

Source data

Extended Data Fig. 10 Proposed model for early-life thymic expansion of microbiota-specific T cells.

At weaning we propose that CX3CR1+ DCs take up intestinal commensal microorganisms (1) and migrate to the thymus (2), where they present bacterial antigens to CD4+ T cells. This induces the expansion of bacteria-specific CD4+T cells (3) that are later exported to peripheral organs (4).

Supplementary information

Reporting Summary

Supplementary Table 1

Normalized gene counts of SFB-specific and non-specific CD4 SP thymocytes as determined by RNA-seq. Normalized gene counts obtained by DESeq.

Supplementary Table 2

Differential Gene Expression between SFB-specific and non-specific CD4 SP thymocytes by RNA-seq.Differential gene expression analyzed by DESeq. Two-tailed P-values generated by the Wald test were corrected for multiple testing using the Benjamini and Hochberg method.

Supplementary Table 3

KEGG Pathways identified as differentially enriched between SFB-specific and non-specific CD4 SP thymocytes. Top 200 differentially expressed genes were analyzed in DAVID v6.8 using KEGG PATHWAY as a reference database. Significantly gene-enriched terms (p<0.05) are shown. One-sided P value was obtained by Fisher’s exact test (EASE score).

Supplementary Table 4

Bacteria-specific T cells from the thymus are functional in distal tissues. Experimental groups for transfer of thymic T cells to RAG-/- recipients.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zegarra-Ruiz, D.F., Kim, D.V., Norwood, K. et al. Thymic development of gut-microbiota-specific T cells. Nature 594, 413–417 (2021). https://doi.org/10.1038/s41586-021-03531-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-03531-1

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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