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:

Mapping the T cell repertoire to a complex gut bacterial community

Abstract

Certain bacterial strains from the microbiome induce a potent, antigen-specific T cell response1,2,3,4,5. However, the specificity of microbiome-induced T cells has not been explored at the strain level across the gut community. Here, we colonize germ-free mice with complex defined communities (roughly 100 bacterial strains) and profile T cell responses to each strain. The pattern of responses suggests that many T cells in the gut repertoire recognize several bacterial strains from the community. We constructed T cell hybridomas from 92 T cell receptor (TCR) clonotypes; by screening every strain in the community against each hybridoma, we find that nearly all the bacteria-specific TCRs show a one-to-many TCR-to-strain relationship, including 13 abundant TCR clonotypes that each recognize 18 Firmicutes. By screening three pooled bacterial genomic libraries, we discover that these 13 clonotypes share a single target: a conserved substrate-binding protein from an ATP-binding cassette transport system. Peripheral regulatory T cells and T helper 17 cells specific for an epitope from this protein are abundant in community-colonized and specific pathogen-free mice. Our work reveals that T cell recognition of commensals is focused on widely conserved, highly expressed cell-surface antigens, opening the door to new therapeutic strategies in which colonist-specific immune responses are rationally altered or redirected.

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: A model system for studying immune modulation by the gut microbiome.
Fig. 2: Strain-by-strain profiling of T cell responses to a complex defined community.
Fig. 3: scRNA-seq and scTCR-seq to identify microbiome-responsive T cell clonotypes.
Fig. 4: Discovery of a conserved Firmicutes antigen.

Similar content being viewed by others

Data availability

The data have been deposited as follows: Cell Ranger out files, https://doi.org/10.5281/zenodo.8008419; raw sequence data: scTCR-seq of T cells from the small intestine of germ-free mice, https://doi.org/10.5281/zenodo.8007653; scTCR-seq of T cells from the small intestine of hCom1 colonized mice, https://doi.org/10.5281/zenodo.8007692; scTCR-seq of T cells from the small intestine of hCom2-colonized mice, https://doi.org/10.5281/zenodo.8007696; scTCR-seq of T cells from the large intestine of germ-free mice, https://doi.org/10.5281/zenodo.8007700; scTCR-seq of T cells from the large intestine of hCom1 colonized mice, https://doi.org/10.5281/zenodo.8007704; scTCR-seq of T cells from the large intestine of hCom2-colonized mice, https://doi.org/10.5281/zenodo.8007709; scRNA-seq of T cells from the small intestine of germ-free mice, https://doi.org/10.5281/zenodo.8007733; scRNA-seq of T cells from the small intestine of hCom1 colonized mice, https://doi.org/10.5281/zenodo.8007735; scRNa-seq of T cells from the small intestine of hCom2-colonized mice, https://doi.org/10.5281/zenodo.8007737; scRNA-seq of T cells from the large intestine of germ-free mice, https://doi.org/10.5281/zenodo.8007740; scRNA-seq of T cells from the large intestine of hCom1 colonized mice, https://doi.org/10.5281/zenodo.8007742; scRNA-seq of T cells from the large intestine of hCom2-colonized mice, https://doi.org/10.5281/zenodo.8007744Source data are provided with this paper.

Code availability

The custom code for scRNA-seq and scTCR-seq analyses is deposited at Zenodo (https://doi.org/10.5281/zenodo.8011206). The following software packages were used: R v.3.6.2 (2019-12-12), platform x86_64-apple-darwin15.6.0 (64-bit); macOS v.10.16; the R packages Seurat v.3.1.5 and scRepertoire v.1.0.0; Microsoft Excel v.16.73; FlowJo (TreeStar) v.10.8.1; Prism 9 for macOS (graphpad) v.9.5.1; PyMOL (Schrödinger, Inc.) v.2.5.

References

  1. 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 

  2. 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 

  3. Linehan, J. L. et al. Non-classical immunity controls microbiota impact on skin immunity and tissue repair. Cell 172, 784–796.e18 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ansaldo, E. et al. Akkermansia muciniphila induces intestinal adaptive immune responses during homeostasis. Science 364, 1179–1184 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ivanov, I. I., Tuganbaev, T., Skelly, A. N. & Honda, K. T cell responses to the microbiota. Annu. Rev. Immunol. 40, 559–587 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Gilbert, J. A. et al. Current understanding of the human microbiome. Nat. Med. 24, 392–400 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Fan, Y. & Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 19, 55–71 (2021).

    Article  CAS  PubMed  Google Scholar 

  8. Smith, M. I. et al. Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science 339, 548–554 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sharon, G. et al. Human gut microbiota from autism spectrum disorder promote behavioral symptoms in mice. Cell 177, 1600–1618.e17 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ridaura, V. K. et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 341, 1241214 (2013).

    Article  PubMed  Google Scholar 

  11. Schaubeck, M. et al. Dysbiotic gut microbiota causes transmissible Crohn’s disease-like ileitis independent of failure in antimicrobial defence. Gut 65, 225–237 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97–103 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91–97 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  14. Matson, V. et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 359, 104–108 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ivanov, I. I. et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139, 485–498 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kullberg, M. C. et al. Bacteria-triggered CD4(+) T regulatory cells suppress Helicobacter hepaticus-induced colitis. J. Exp. Med. 196, 505–515 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chai, J. N. et al. Helicobacter species are potent drivers of colonic T cell responses in homeostasis and inflammation. Sci. Immunol. 2, eaal5068 (2017).

  18. Palm, N. W. et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 158, 1000–1010 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Fagarasan, S., Kawamoto, S., Kanagawa, O. & Suzuki, K. Adaptive immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis. Annu. Rev. Immunol. 28, 243–273 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Belkaid, Y. & Hand, T. W. Role of the microbiota in immunity and inflammation. Cell 157, 121–141 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Surana, N. K. & Kasper, D. L. Moving beyond microbiome-wide associations to causal microbe identification. Nature 552, 244–247 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  23. Tanoue, T. et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 565, 600–605 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  25. Geva-Zatorsky, N. et al. Mining the human gut microbiota for immunomodulatory organisms. Cell 168, 928–943.e11 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cheng, A. G. et al. Design, construction, and in vivo augmentation of a complex gut microbiome. Cell 185, 3617–3636.e19 (2022).

    Article  CAS  PubMed  Google Scholar 

  27. Moran, A. E. et al. T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. J. Exp. Med. 208, 1279–1289 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ashouri, J. F. & Weiss, A. Endogenous nur77 is a specific indicator of antigen receptor signaling in human T and B cells. J. Immunol. 198, 657–668 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Kiner, E. et al. Gut CD4+ T cell phenotypes are a continuum molded by microbes, not by TH archetypes. Nat. Immunol. 22, 216–228 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ise, W. et al. CTLA-4 suppresses the pathogenicity of self antigen-specific T cells by cell-intrinsic and cell-extrinsic mechanisms. Nat. Immunol. 11, 129–135 (2010).

    Article  CAS  PubMed  Google Scholar 

  31. Wegorzewska, M. M. et al. Diet modulates colonic T cell responses by regulating the expression of a Bacteroides thetaiotaomicron antigen. Sci. Immunol. 4, (2019).

  32. Bousbaine, D. et al. A conserved Bacteroidetes antigen induces anti-inflammatory intestinal T lymphocytes. Science 377, 660–666 (2022).

  33. Kuczma, M. P. et al. Commensal epitopes drive differentiation of colonic Tregs. Sci. Adv. 6, eaaz3186 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cong, Y., Feng, T., Fujihashi, K., Schoeb, T. R. & Elson, C. O. A dominant, coordinated T regulatory cell-IgA response to the intestinal microbiota. Proc. Natl Acad. Sci. USA 106, 19256–19261 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lee, S.-J. et al. Temporal expression of bacterial proteins instructs host CD4 T cell expansion and Th17 development. PLoS Pathog. 8, e1002499 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. van der Heide, T. & Poolman, B. ABC transporters: one, two or four extracytoplasmic substrate-binding sites? EMBO Rep. 3, 938–943 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Brautigam, C. A., Deka, R. K., Liu, W. Z. & Norgard, M. V. The Tp0684 (MglB-2) lipoprotein of Treponema pallidum: a glucose-binding protein with divergent topology. PLoS ONE 11, e0161022 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Mehta, R. S. et al. Stability of the human faecal microbiome in a cohort of adult men. Nat. Microbiol. 3, 347–355 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Spindler, M. P. et al. Human gut microbiota stimulate defined innate immune responses that vary from phylum to strain. Cell Host Microbe 30, 1481–1498.e5 (2022).

    Article  CAS  PubMed  Google Scholar 

  40. Miragaia, R. J. et al. Single-cell transcriptomics of regulatory T cells reveals trajectories of tissue adaptation. Immunity 50, 493–504.e7 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Muschaweck, M. et al. Cognate recognition of microbial antigens defines constricted CD4+ T cell receptor repertoires in the inflamed colon. Immunity 54, 2565–2577.e6 (2021).

    Article  CAS  PubMed  Google Scholar 

  42. Perez-Muñoz, M. E., Joglekar, P., Shen, Y.-J., Chang, K. Y. & Peterson, D. A. Identification and phylogeny of the first T cell epitope identified from a human gut bacteroides species. PLoS ONE 10, e0144382 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Bunker, J. J. et al. Natural polyreactive IgA antibodies coat the intestinal microbiota. Science 358, eaan6619 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lindner, C. et al. Diversification of memory B cells drives the continuous adaptation of secretory antibodies to gut microbiota. Nat. Immunol. 16, 880–888 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Rollenske, T. et al. Parallelism of intestinal secretory IgA shapes functional microbial fitness. Nature 598, 657–661 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  46. Cameroni, E. et al. Broadly neutralizing antibodies overcome SARS-CoV-2 Omicron antigenic shift. Nature 602, 664–670 (2022).

    Article  ADS  CAS  PubMed  Google Scholar 

  47. Burton, D. R. & Hangartner, L. Broadly neutralizing antibodies to HIV and their role in vaccine design. Annu. Rev. Immunol. 34, 635–659 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chen, Y. E. et al. Engineered skin bacteria induce antitumor T cell responses against melanoma. Science 380, 203–210 (2023).

  49. Dobson, C. S. et al. Antigen identification and high-throughput interaction mapping by reprogramming viral entry. Nat. Methods 9, 449–460 (2022).

  50. Yu, B. et al. Engineered cell entry links receptor biology with single-cell genomics. Cell 185, 4904–4920.e22 (2021).

  51. Curran, M. A. & Allison, J. P. Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer Res. 69, 7747–7755 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Borcherding, N., Bormann, N. L. & Kraus, G. scRepertoire: an R-based toolkit for single-cell immune receptor analysis. F1000Res. 9, 47 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sanderson, S., Campbell, D. J. & Shastri, N. Identification of a CD4+ T cell-stimulating antigen of pathogenic bacteria by expression cloning. J. Exp. Med. 182, 1751–1757 (1995).

    Article  CAS  PubMed  Google Scholar 

  55. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  56. Evans, R. et al. Protein complex prediction with AlphaFold-Multimer. Preprint at bioRxiv https://doi.org/10.1101/2021.10.04.463034 (2021).

  57. Mirdita, M. et al. ColabFold: making protein folding accessible to all. Nat. Methods 19, 679–682 (2022).

  58. Centanni, M., Sims, I. M., Bell, T. J., Biswas, A. & Tannock, G. W. Sharing a β-glucan meal: transcriptomic eavesdropping on a Bacteroides ovatus-Subdoligranulum variabile-Hungatella hathewayi consortium. Appl. Environ. Microbiol. 86, e01651-20 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  59. Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are deeply indebted to D. Bousbaine and other members of the Fischbach Group for helpful suggestions and comments on the manuscript. We thank H. Takayanagi, S. Sawa, R. Muro, N. A. Bracey, M. M. Davis and members of their laboratories for useful discussions. We are grateful to the Stanford Shared FACS Facility for access to flow cytometry and the Stanford Genomics Facility for constructing and sequencing libraries for scRNA-seq and scTCR-seq experiments, and the Harvard Faculty of Arts & Sciences Research Computing Cluster for computational resources. This work was supported by the Stanford Microbiome Therapies Initiative, the Human Frontier Science Program grant no. LT000493/2018-L (K.N.), a Fellowship from the Astellas Foundation for Research on Metabolic Disorders (K.N.), a research grant from Kanae Fundation for the Promotion of Medical Science (K.N.), NIH grant no. K99AI173524 (K.N.), an HHMI-LSRF Award (J.E.B.), a fellowship from the Kwanjeong Educational Foundation (M.B.), the Howard Hughes Medical Institute (E.P.B.), the Alan T. Waterman Award from the National Science Foundation (E.P.B., grant no. CHE-20380529), an HHMI-Simons Faculty Scholar Award (M.A.F.), the Leona M. and Harry B. Helmsley Charitable Trust (M.A.F.), NIH grant no. DK110174 (M.A.F.), the Chan Zuckerberg Biohub (M.A.F.), Stand Up to Cancer (M.A.F.) and the MAC3 Impact Philanthropies (M.A.F.).

Author information

Authors and Affiliations

Authors

Contributions

K.N., M.B., E.P.B. and M.A.F. conceived and designed the experiments. K.N., A.Z., K.A., M.B., J.E.B., A.W., S.J., X.M., A.G.C., M.W., S.H., A.D., J.J.M. and P.M. performed the experiments. K.N., M.B. and M.A.F. analysed data and wrote the manuscript. M.B., E.S.S. and E.P.B. edited the manuscript. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Michael A. Fischbach.

Ethics declarations

Competing interests

Stanford University and the Chan Zuckerberg Biohub have patents pending for microbiome technologies on which the authors are co-inventors. M.A.F. is a cofounder and director of Federation Bio and Kelonia, a cofounder of Revolution Medicines and a member of the scientific advisory boards of the Chan Zuckerberg Initiative and NGM Bio. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature thanks Jeremiah Faith, Kylie James 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 figures and tables

Extended Data Fig. 1 T cell profiling for hCom1d- and hCom2d-colonized mice.

a,b, hCom1d or hCom2d were used to colonize germ-free C57BL/6 mice by oral gavage. Mice were housed for two weeks before sacrifice. Intestinal immune cells were extracted, stimulated by PMA/ionomycin and analyzed by flow cytometry. Th cell subtypes, as a percentage of the total Th cell pool, were analyzed in the large intestine (a) or in the small intestine (b). See Supplementary Fig. 1b for the gating strategy. Statistical significance was assessed using a one-way ANOVA (NS > 0.05; **p < 0.01; ***p < 0.001). Data shown are mean ± standard deviations. n = 8, 8, 4 mice per group from 2 independent experiments (a). n = 4 mice per group from one experiment (b).

Source Data

Extended Data Fig. 2 Introduction of a gut bacterial community to germ-free mice by horizontal transfer.

a, Metagenomic analysis of germ-free mice colonized with hCom2d by oral gavage or cohousing (horizontal transfer, HT). Faecal samples were collected 2 weeks after colonization and subjected to metagenomic sequencing; the resulting data were analyzed by NinjaMap to measure the composition of each community. The average of 4 mice per group is displayed above. Each dot is an individual strain; the collection of dots in a column represents the community at a single time point. Strains are coloured according to their rank-order abundance in the oral gavage sample. Relative abundances in the oral gavage and HT samples are highly correlated, indicating that coprophagy (horizontal transfer) results in a similar community architecture to that of oral gavage. b, Intestinal T cells from mice colonized with hCom2d by oral gavage and HT show similar phenotypes. Mice were sacrificed after two weeks of colonization. Immune cells were isolated from the large intestine, stimulated by PMA/ionomycin and analyzed by flow cytometry. See Supplementary Fig. 1b for the gating strategy. Statistical significance was assessed using a two-sided t-test (NS > 0.05). Data shown are mean ± standard deviations. n = 6 mice per group from one experiment.

Source Data

Extended Data Fig. 3 The mixed lymphocyte assay detects TCR activation while preserving T cell phenotype.

a-c, We colonized germ-free mice with hCom1d, waited two weeks, and then sacrificed the mice. Immune cells from the colon were co-cultured for 4 h with three non-community bacterial strains (Bacillus cereus, Bacillus subtilis, or Staphylococcus epidermidis) or two strains from hCom1d (Tyzzerella nexilis and Clostridium bolteae). For antigen presentation, we used either MHCII+ wild-type DCs, MHCII-deficient DCs or a no DC control. pTreg (Helios- Foxp3+) cells and Th17 (RORgt+ Foxp3-) cells were analyzed by flow cytometry. a, Co-culture with strains and wild-type DCs doesn’t affect the number of pTreg cells and Th17 cells. b,c, Nur77 expression is upregulated when T cells were cocultured with strains in hCom1d and wild-type DCs in pTreg cells (b) and in Th17 cells (c). p-values were calculated by comparison to PBS treatment as a negative control using a one-way ANOVA (NS > 0.05; **p < 0.01; ***p < 0.001). Data shown are mean ± standard deviations. n = 4 mice per group from one experiment.

Source Data

Extended Data Fig. 4 Analysis of scRNA-seq data by unbiased clustering.

a, Uniform manifold approximation and projection (UMAP) plot of all the cells. The data is generated by merging three groups: hCom1d-colonized, hCom2d-colonized, and germ-free mice. Cells were clustered into 25 groups using the FindClusters function of the Seurat R package. b–d, Gene expression profiling of cell clusters. To investigate the identity of each cell cluster, expression levels of cell subset markers were visualized by dot plots (b), feature plots (c) and violin plots (d) using Seurat.

Source Data

Extended Data Fig. 5 scRNA-seq analysis of immune modulation by synthetic community colonization.

a, Left panel: Uniform manifold approximation and projection (UMAP) plot. These data represent three merged samples: hCom1d-colonized, hCom2d-colonized and germ-free mice. Right panel: Frequency of TCR clonotypes on the UMAP plot. Expanded TCRs (red) represent clonotypes observed in more than five cells, multiple (orange) are clonotypes found in 2–5 cells, and single (light blue) were seen in only one cell. Most of the expanded TCR clonotypes have an expression profile consistent with effector T cells, whereas naïve T cells are rich in unique (that is, non- expanded) TCR clonotypes. b, UMAP plot of intestinal immune cell clusters in each colonization condition. Immune cells were isolated from the large and small intestine from three groups of mice: hCom1d-colonized, hCom2d-colonized, and germ-free. c, Analysis of the frequency of T cell subsets in each group. The percentage of each T cell subset on the UMAP plot was calculated by the Seurat R package; fold changes compared to GF mice are shown. Colonization of germ-free mice with hCom1d and hCom2d increased pTreg, Th17, Fr4 Th and other effector T cells in the large intestine, and Th17 and other effector T cells in the small intestine. d, Analysis of expanded TCR clonotypes in each sample. Each dot represents one TCR clonotype found in multiple T cells (red, shared between effector T cells and pTreg cells; grey, effector T cell; black, pTreg). e, Differentially expressed genes in T cell subsets upon colonization with hCom1d and hCom2d. The FindMarkers function of Seurat was used to find differentially expressed genes. The two-sided non-parametric Wilcoxon rank sum test was used to calculate the adjusted p-value. White bars show mean values. Each dot represents one cell. ***p < 0.001. f, Criteria used to select TCR clonotypes for making hybridoma cells. Red genes: Upregulated by hCom1d and hCom2d colonization. Black genes: T cell subset markers.

Source Data

Extended Data Fig. 6 Reactivity of TCR hybridomas against reported commensal epitopes.

a, The list of previously reported commensal epitopes tested in this experiment. Epitopes were pooled into sets of 6-7 for testing the ability to stimulate TCR hybridomas. b, Previously reported commensal epitopes were co-cultured with TCR hybridomas and dendritic cells. The SFB-specific 7B8 TCR transgenic T cells, a positive control, showed a response to Pool1, which includes the SFB antigen SFB3340. The 92 TCR hybridomas generated in this work were not responsive to any of the previously reported commensal epitopes.

Source Data

Extended Data Fig. 7 Metagenomic analysis for hCom1d and hCom2d strains in the colonization of the mouse intestine.

a,b, Each dot is an individual strain; the collection of dots in a column represents the community at a single time point in mice colonized by hCom1d (a) or hCom2d (b). Strains are colored according to their rank-order relative abundance. Strain names colored red harbor the conserved substrate-binding protein (SBP). Germ-free mice were colonized with synthetic communities, and fecal pellets were collected two weeks after community colonization. The results are an average of 5 mice.

Source Data

Extended Data Fig. 8 Identification of the minimal antigen epitopes of SBP using a truncated peptide library.

a,b, Prediction of signal sequences and subcellular localization of SBP. SignalP-5.0 (a) and PRED-LIPO (b) predict that the SBP has a lipoprotein signal peptide. c, BLAST search for the antigenic epitopes of SBP in strains from hCom1d and hCom2d. 19 strains were found to harbor homologs of the SBP. d, To search for the minimal antigenic epitope in SBP, a library of truncated peptides was synthesized. 13 Firmicutes-reactive TCR hybridomas were mixed and co-cultured with truncated peptides and dendritic cells. The degree of TCR stimulation was estimated by assaying the concentration of IL-2 in the culture supernatant by ELISA. Truncated peptides containing the 9-mer YDAFAINMV stimulated the mixed TCR hybridomas.

Source Data

Extended Data Fig. 9 Discovery of an N-terminal epitope from SBP.

a, Identification of the N-terminal epitope SBP76-84. N-terminal SBP peptides from strains from hCom1d and hCom2d were synthesized and co-cultured with the T cell hybridomas. Truncated peptides were also tested to identify the minimal epitope. 7 of the 13 TCRs were responsive to the synthetic peptides. The remaining 6 were stimulated by C-terminal peptide SBP405-413 as shown in Fig. 4f. There is a strong correlation between the reactivity of TCRs and the sequences of TCR CD3 regions. b, The T cell response to SBP is conserved in different murine settings. We colonized: (1) germ-free C57BL/6 mice with hCom2d by co-housing (horizontal transfer recipient), (2) germ-free C57BL/6 mice with a human fecal community (humanized), (3) germ-free C57BL/6 mice with oral gavage of hCom2d under a condition of a high-fat diet (HFD), and (4) germ-free Swiss Webster mice (SW) with oral gavage of hCom2d. After two weeks, intestinal immune cells were isolated and cocultured with a mix of SBP76-84 and SBP405-413, or PBS as a negative control, using dendritic cells for antigen presentation. Nur77 expression in pTreg or Th17 cells was analyzed by FACS to monitor TCR stimulation (see Supplementary Fig. 1a, c for the gating strategy). SBP-specific T cells were detected in pTreg and Th17 from HT recipient, humanized, and HFD groups, while T cells from SW mice failed to respond to SBP76-84 and SBP405-413. c, We hypothesized that T cells from SW mice recognize a distinct epitope in SBP because of the difference in MHC haplotype between C57BL/6 and SW mice. To test this hypothesis, we co-cultured immune cells from hCom2d-colonized SW mice with a mixture of peptides that tile the whole SBP. We detected SBP-specific pTreg and Th17 cells in hCom2-colonized SW mice. This suggests that SBP-specific T cell induction is preserved across different genetic backgrounds of mice, but—as expected—the epitope recognized is distinct because of the difference in MHC haplotype. p-values were calculated using a two-sided t-test by comparison to PBS treatment as a negative control. *p = 0.05. **p = 0.01. ***p = 0.005. NS > 0.05. Data shown are mean ± standard deviations. n = 8, 8, 14, 8 mice per group from 2 independent experiments (b). n = 15 mice per group from one experiment (c).

Source Data

Extended Data Fig. 10 Identification of TPRL as an antigen from Bacteroides species.

a, To search for the antigenic epitope in TPRL from Bacteroides eggerthii, a library of truncated peptides was synthesized. 4 Bacteroides-reactive TCR hybridomas were mixed and co-cultured with truncated peptides and dendritic cells. The degree of TCR stimulation was estimated by assaying the concentration of IL-2 in the culture supernatant by ELISA. TPRL29-53 (Peptide 3, SDYFTVTPQVLEAVGGKVPATINGK) stimulated the mixed TCR hybridomas. b, We found that TCR H2-11, H2-30, and H1-14 are reactive to Peptide 3 from TPRL by coculturing each Bacteroides-reactive TCR hybridoma with Peptides 2 and 3. c, Results of a BLAST search using the antigenic epitope TPRL29-53 as a query; the results shown are from strains in hCom1d and hCom2d. d, Predicted crystal structure of the TPRL from AlphaFold2. The TPRL29-53 epitope, shown in red, lies within a beta-sheet in the N-terminal domain. e, Induction of TPRL-specific T cells in vivo. Germ-free C57BL/6 mice were colonized with hCom2d. After two weeks, intestinal T cells were isolated and cocultured with TPRL29-53 and dendritic cells. Nur77 expression in T cell subsets was analyzed by FACS to monitor TCR stimulation (see Extended Data Fig. 1a, c for the gating strategy). In hCom1d and hCom2d-colonized mice, Th17 and pTreg cells showed an antigen-specific response to SBP. p-values were calculated using a two-sided t-test by comparison to PBS treatment as a negative control. *p < 0.05. Data shown are mean ± standard deviations. n = 5, 8 mice per group from one experiment.

Source Data

Extended Data Fig. 11 T cell targets are highly expressed in vitro and in vivo.

a, The SBP is highly expressed in in vitro transcriptomic data from two species in our community: Clostridium bolteae and Clostridium hathewayi. In C. bolteae, it is the 10th most highly expressed gene out of 5,982, and in C. hathewayi, it is the 4th most highly expressed gene out of 6,712. The expression level was normalized to transcripts per million (TPM). b, The SBP and TPRL are highly expressed in vivo. Reads were recruited from 378 metatranscriptomic samples38 and mapped to the whole genomes of select SBP and TPRL-encoding strains in our community (Tyzzerella nexilis DSM 1787 and Clostridium bolteae DSM 15670 (SBP); Bacteroides stercoris ATCC 43183, Bacteroides eggerthii DSM 20697, and Bacteroides cellulosilyticus DSM 14838 (TPRL)). To account for gene copy number variation, we measured expression as the ratio between RNA and DNA level from paired metagenomic and metatranscriptomic samples. In all cases, we found that the protein is very highly expressed in vivo.

Source Data

Extended Data Fig. 12 SBP and TPRL are present in human stool samples.

We tested whether SBP and TPRL exist at a detectable level in human stool samples. We cocultured SBP- or TPRL-reactive TCR hybridomas with: (1) PBS as a negative control, (2) a mix of SBP and TPRL epitopes as a positive control, (3) heat-treated hCom1d, (4) heat-treated SPF mouse fecal pellets, 5) heat-treated mouse fecal pellets from Jackson, (5) 6 human fecal communities. For antigen presentation, we used MHCII+ wild-type DCs, or MHCII-deficient DCs as a negative control. IL-2 concentration was measured as a readout for TCR stimulation. We found that 6/6 human fecal communities restimulate the SBP405-413 and SBP76-84-specific hybridomas and 5/6 restimulate the TPRL29-53-specific hybridomas. These data suggest that the SBP and TPRL are expressed by human gut isolates under native conditions.

Source Data

Supplementary information

Supplementary Figures

This file contains Supplementary Figs. 1–3. Supplementary Fig. 1 Gating strategy for T cell in flow cytometry analysis. Supplementary Fig. 2 Establishment of the mixed lymphocyte assay. Supplementary Fig. 3 Representative flow cytometry images in Fig. 2. See PDF for full legends.

Reporting Summary

Supplementary Table 1

List of strains in hCom1d and hCom2d. Strains in hCom1d and hCom2d and their respective growth media are shown.

Supplementary Table 2

TCR clonotypes chosen for hybridoma cell lines. Information on TCR clonotypes picked up for hybridoma cells is described.

Supplementary Table 3

Plasmid and gBlock sequences. Sequences of plasmids and gBlocks used in this study are listed.

Supplementary Table 4

P values. The P values from this study are listed.

Supplementary Data

Source data for Supplementary Figs.

Source data

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

Nagashima, K., Zhao, A., Atabakhsh, K. et al. Mapping the T cell repertoire to a complex gut bacterial community. Nature 621, 162–170 (2023). https://doi.org/10.1038/s41586-023-06431-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06431-8

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 Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

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