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:

The endogenous repertoire harbors self-reactive CD4+ T cell clones that adopt a follicular helper T cell-like phenotype at steady state

Abstract

The T cell repertoire of healthy mice and humans harbors self-reactive CD4+ conventional T (Tconv) cells capable of inducing autoimmunity. Using T cell receptor profiling paired with in vivo clonal analysis of T cell differentiation, we identified Tconv cell clones that are recurrently enriched in non-lymphoid organs following ablation of Foxp3+ regulatory T (Treg) cells. A subset of these clones was highly proliferative in the lymphoid organs at steady state and exhibited overt reactivity to self-ligands displayed by dendritic cells, yet were not purged by clonal deletion. These clones spontaneously adopted numerous hallmarks of follicular helper T (TFH) cells, including expression of Bcl6 and PD-1, exhibited an elevated propensity to localize within B cell follicles at steady state, and produced interferon-γ in non-lymphoid organs following sustained Treg cell depletion. Our work identifies a naturally occurring population of self-reactive TFH-like cells and delineates a previously unappreciated fate for self-specific Tconv cells.

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: Recurrent CD4+ Tconv clones are detected in non-lymphoid organs of Treg cell-depleted mice.
Fig. 2: Multiple recurrent prostate-infiltrating CD4+ Tconv clones exhibit hallmarks of steady-state activation and reactivity to MHC-II-restricted self-ligands.
Fig. 3: Group 3 clones express common hallmarks of TFH cells.
Fig. 4: T cells expressing Group 3 TCRs do not produce common helper T effector cytokines or promote spontaneous germinal center formation.
Fig. 5: Following Treg cell ablation, Group 3 clones are enriched in non-lymphoid organs and produce IFN-γ.
Fig. 6: In the thymus, Group 3 Tconv clones display hallmarks of elevated self-reactivity.
Fig. 7: The endogenous CD4+ Tconv repertoire harbors T cells displaying hallmarks of Group 3 clones.

Similar content being viewed by others

Data availability

RNA-seq data are available at the NIH Gene Expression Omnibus under accession code GSE190127. All other source data, including TCR sequencing data, are provided with this manuscript. Source data are provided with this paper.

References

  1. 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 

  2. Hogquist, K. A. & Jameson, S. C. The self-obsession of T cells: how TCR signaling thresholds affect fate ‘decisions’ and effector function. Nat. Immunol. 15, 815–823 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bouneaud, C., Kourilsky, P. & Bousso, P. Impact of negative selection on the T cell repertoire reactive to a self-peptide: a large fraction of T cell clones escapes clonal deletion. Immunity 13, 829–840 (2000).

    Article  CAS  PubMed  Google Scholar 

  4. Zehn, D. & Bevan, M. J. T cells with low avidity for a tissue-restricted antigen routinely evade central and peripheral tolerance and cause autoimmunity. Immunity 25, 261–270 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Yu, W. et al. Clonal deletion prunes but does not eliminate self-specific αβ CD8+ T lymphocytes. Immunity 42, 929–941 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kim, J. M., Rasmussen, J. P. & Rudensky, A. Y. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 8, 191–197 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Lahl, K. et al. Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease. J. Exp. Med. 204, 57–63 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chinen, T., Volchkov, P. Y., Chervonsky, A. V. & Rudensky, A. Y. A critical role for regulatory T cell-mediated control of inflammation in the absence of commensal microbiota. J. Exp. Med. 207, 2323–2330 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Yi, J. et al. Unregulated antigen-presenting cell activation by T cells breaks self tolerance. Proc. Natl Acad. Sci. USA 116, 1007–1016 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Fernando, M. M. A. et al. Defining the role of the MHC in autoimmunity: a review and pooled analysis. PLoS Genet. 4, e1000024 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Sakaguchi, S., Sakaguchi, N., Asano, M., Itoh, M. & Toda, M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor α-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J. Immunol. 155, 1151–1164 (1995).

    Article  CAS  PubMed  Google Scholar 

  12. Morrissey, P. J., Charrier, K., Braddy, S., Liggitt, D. & Watson, J. D. CD4+ T cells that express high levels of CD45RB induce wasting disease when transferred into congenic severe combined immunodeficient mice. Disease development is prevented by cotransfer of purified CD4+ T cells. J. Exp. Med. 178, 237–244 (1993).

    Article  CAS  PubMed  Google Scholar 

  13. Powrie, F., Leach, M. W., Mauze, S., Caddie, L. B. & Coffman, R. L. Phenotypically distinct subsets of CD4+ T cells induce or protect from chronic intestinal inflammation in c. B-17 scid mice. Int. Immunol. 5, 1461–1471 (1993).

    Article  CAS  PubMed  Google Scholar 

  14. Kim, H. J., Verbinnen, B., Tang, X., Lu, L. & Cantor, H. Inhibition of follicular T-helper cells by CD8+ regulatory T cells is essential for self tolerance. Nature 467, 328–332 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Richards, D. M. et al. The contained self-reactive peripheral T cell repertoire: size, diversity, and cellular composition. J. Immunol. 195, 2067–2079 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. Kawabe, T. et al. Memory-phenotype CD4+ T cells spontaneously generated under steady-state conditions exert innate TH1-like effector function. Sci. Immunol. 2, eaam9304 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Levine, A. G. et al. Stability and function of regulatory T cells expressing the transcription factor T-bet. Nature 546, 421–425 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kawabe, T. et al. Requirements for the differentiation of innate T-bet high memory-phenotype CD4+ T lymphocytes under steady state. Nat. Commun. 11, 3366 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kalekar, L. A. et al. CD4+ T cell anergy prevents autoimmunity and generates regulatory T cell precursors. Nat. Immunol. 17, 304–314 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Malchow, S. et al. Aire-dependent thymic development of tumor-associated regulatory T cells. Science 339, 1219–1224 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Malchow, S. et al. Aire enforces immune tolerance by directing autoreactive T cells into the regulatory T cell lineage. Immunity 44, 1102–1113 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sant’Angelo, D. B. et al. The imprint of intrathymic self-peptides on the mature T cell receptor repertoire. Immunity 7, 517–524 (1997).

    Article  PubMed  Google Scholar 

  23. Hsieh, C. S. et al. Recognition of the peripheral self by naturally arising CD25+CD4+ T cell receptors. Immunity 21, 267–277 (2004).

    Article  CAS  PubMed  Google Scholar 

  24. Miller, C. H. et al. Eomes identifies thymic precursors of self-specific memory-phenotype CD8+ T cells. Nat. Immunol. 21, 567–577 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. McDonald, B. D., Bunker, J. J., Erickson, S. A., Oh-Hora, M. & Bendelac, A. Crossreactive αβ T cell receptors are the predominant targets of thymocyte negative selection. Immunity 43, 859–869 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Miyazaki, T. et al. Mice lacking H2-M complexes, enigmatic elements of the MHC class II peptide-loading pathway. Cell 84, 531–541 (1996).

    Article  CAS  PubMed  Google Scholar 

  27. Subramanian, A. et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Nurieva, R. I. et al. Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages. Immunity 29, 138–149 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wei, G. et al. Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity 30, 155–167 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Johnston, R. J. et al. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science 325, 1006–1010 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yusuf, I. et al. Germinal center T follicular helper cell IL-4 production is dependent on signaling lymphocytic activation molecule receptor (CD150). J. Immunol. 185, 190–202 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Kitano, M. et al. Bcl6 protein expression shapes pre-germinal center B cell dynamics and follicular helper T cell heterogeneity. Immunity 34, 961–972 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Liu, X. et al. Bcl6 expression specifies the T follicular helper cell program in vivo. J. Exp. Med. 209, 1841–1852 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Samstein, R. M. et al. Foxp3 exploits a pre-existent enhancer landscape for regulatory T cell lineage specification. Cell 151, 153–166 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hale, J. S. et al. Distinct memory CD4+ T cells with commitment to T follicular helper- and T helper 1-cell lineages are generated after acute viral infection. Immunity 38, 805–817 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Crotty, S. T follicular helper cell differentiation, function, and roles in disease. Immunity 41, 529–542 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Vinuesa, C. G., Linterman, M. A., Yu, D. & Maclennan, I. C. M. Follicular helper T cells. Annu. Rev. Immunol. 34, 335–368 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Crotty, S. T follicular helper cell biology: a decade of discovery and diseases. Immunity 50, 1132–1148 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Xu, W. et al. The transcription factor Tox2 drives T follicular helper cell development via regulating chromatin accessibility. Immunity 51, 826–839 (2019).

    Article  CAS  PubMed  Google Scholar 

  40. Nurieva, R. I. et al. Bcl6 mediates the development of T follicular helper cells. Science 325, 1001–1005 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. McDonald, B. D., Bunker, J. J., Ishizuka, I. E., Jabri, B. & Bendelac, A. Elevated T cell receptor signaling identifies a thymic precursor to the TCRαβ+CD4CD8β intraepithelial lymphocyte lineage. Immunity 41, 219–229 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Baldwin, T., Sandau, M. M., Jameson, S. C. & Hogquist, K. A. The timing of TCRα expression critically influences T cell development and selection. J. Exp. Med. 202, 111–121 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Klawon, D. E. J. et al. Altered selection on a single self-ligand promotes susceptibility to organ-specific T cell infiltration. J. Exp. Med. 218, e20200701 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hogquist, K. A. Assays of thymic selection. Fetal thymus organ culture and in vitro thymocyte dulling assay. Methods Mol. Biol. 156, 219–232 (2001).

    CAS  PubMed  Google Scholar 

  45. Azzam, H. S. et al. CD5 expression is developmentally regulated by T cell receptor (TCR) signals and TCR avidity. J. Exp. Med. 188, 2301–2311 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Bautista, J. L. et al. Intraclonal competition limits the fate determination of regulatory T cells in the thymus. Nat. Immunol. 10, 610–617 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Leung, M. W. L., Shen, S. & Lafaille, J. J. TCR-dependent differentiation of thymic Foxp3+ cells is limited to small clonal sizes. J. Exp. Med. 206, 2121–2130 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Alonso, R. et al. Induction of anergic or regulatory tumor-specific CD4+ T cells in the tumor-draining lymph node. Nat. Commun. 9, 2113 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Simpson, N. et al. Expansion of circulating T cells resembling follicular helper T cells is a fixed phenotype that identifies a subset of severe systemic lupus erythematosus. Arthritis Rheum. 62, 234–244 (2010).

    Article  CAS  PubMed  Google Scholar 

  50. Liu, R. et al. A regulatory effect of IL-21 on T follicular helper-like cell and B cell in rheumatoid arthritis. Arthritis Res. Ther. 14, R255 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhu, C. et al. Increased frequency of follicular helper T cells in patients with autoimmune thyroid disease. J. Clin. Endocrinol. Metab. 97, 943–950 (2012).

    Article  CAS  PubMed  Google Scholar 

  52. Choi, J. Y. et al. Circulating follicular helper-like T cells in systemic lupus erythematosus: association with disease activity. Arthritis Rheumatol. 67, 988–999 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Rathmell, J. C. et al. CD95 (Fas)-dependent elimination of self-reactive B cells upon interaction with CD4+ T cells. Nature 376, 181–184 (1995).

    Article  CAS  PubMed  Google Scholar 

  54. Rathmell, J. C., Townsend, S. E., Xu, J. C., Flavell, R. A. & Goodnow, C. C. Expansion or elimination of B cells in vivo: dual roles for CD40- and Fas (CD95)-ligands modulated by the B cell antigen receptor. Cell 87, 319–329 (1996).

    Article  CAS  PubMed  Google Scholar 

  55. Di Yu et al. Roquin represses autoimmunity by limiting inducible T-cell co-stimulator messenger RNA. Nature 450, 299–303 (2007).

    Article  CAS  PubMed  Google Scholar 

  56. Wing, J. B., Ise, W., Kurosaki, T. & Sakaguchi, S. Regulatory T cells control antigen-specific expansion of Tfh cell number and humoral immune responses via the coreceptor CTLA-4. Immunity 41, 1013–1025 (2014).

    Article  CAS  PubMed  Google Scholar 

  57. Leonard, J. D. et al. Identification of natural regulatory T cell epitopes reveals convergence on a dominant autoantigen. Immunity 47, 107–117 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. 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 

  59. Kennedy, D. E. et al. Novel specialized cell state and spatial compartments within the germinal center. Nat. Immunol. 21, 660–670 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  61. Hamaguchi, Y. et al. The peritoneal cavity provides a protective niche for B1 and conventional B lymphocytes during anti-CD20 immunotherapy in mice. J. Immunol. 174, 4389–4399 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  63. Liao, Y., Smyth, G. K. & Shi, W. FeatureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  64. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. McCarthy, D. J., Chen, Y. & Smyth, G. K. Differential expression analysis of multifactor RNA-seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Google Scholar 

  67. Haimon, Z. et al. Cognate microglia–T cell interactions shape the functional regulatory T cell pool in experimental autoimmune encephalomyelitis pathology. Nat. Immunol. 23, 1749–1762 (2022).

    Article  CAS  PubMed  Google Scholar 

  68. Painter, M. W. et al. Transcriptomes of the B and T lineages compared by multiplatform microarray profiling. J. Immunol. 186, 3047–3057 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was funded by the following sources (to P.A.S.): R01-CA160371, R01-AI110507, U01-AI154560, a Cancer Research Institute Investigator Award, and the University of Chicago Comprehensive Cancer Center. V.L. was supported by a National Institutes of Health (NIH)/National Cancer Institute (NCI) F30 predoctoral fellowship (F30-CA217109). D.R. was supported by an NIH/NCI F30 predoctoral fellowship (F30-CA247264). C.H.M. was supported by an NIH/NCI F30 predoctoral fellowship (F30-CA236061). V.L, D.R., and C.H.M. were supported by the University of Chicago Medical Scientist Training Program (T32-GM007281). J.L.C, M.T.W., and D.E.J.K were supported by T32-AI007090. M.M-C was supported in part by the University of Illinois Center for Clinical and Translational Science (NCATS grant UL1TR002003). N.D.S. was supported by Memorial Sloan Kettering Cancer Center (support grant P30-CA008748). This work was also supported by R01-AI143778 and R01-AI150860 awarded to M.R.C. Flow cytometry and cell sorting were performed at the Cytometry and Antibody Technology Facility at University of Chicago (RRID SCR_017760), which receives financial support from the Cancer Center Support grant (P30-CA014599).

Author information

Authors and Affiliations

Authors

Contributions

V.L., D.R., and P.A.S. conceived the project, designed experiments, and performed data analysis. N.G. designed and performed experiments and performed data analysis. V.L., D.R., S.Z., J.L.C., M.T.W., C.H.M., D.E.J.K., M.H.S., and J.A. performed the experiments. D.E.K. and M.M.-C. performed computational and statistical analysis of RNA-seq data. N.D.S. performed computational and statistical analysis of TCR sequence data. M.R.C. contributed to experimental design and data interpretation. P.A.S., V.L., and D.R. wrote the manuscript.

Corresponding author

Correspondence to Peter A. Savage.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Shohei Hori, Ellen Robey, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey, in collaboration with the Nature Immunology team.

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 Recurrent CD4+ Tconv clones detected in the prostate of Treg cell-depleted mice are found at low frequencies amongst Tconv and Treg subsets of other reference datasets.

Heat map displaying the 20 TCRα sequences from Fig. 1C and their corresponding log10 frequency in six groups of samples, as indicated. From left to right, these groups are Tconv cells from the prostate of Treg-depleted Foxp3DTR-EGFP males (n = 5), Tconv cells from the salivary glands of Treg-depleted Foxp3DTR-EGFP males (n = 5), Tconv cells from the prostate of untreated Aire−/− males (n = 9), Tconv cells from the prostate of untreated TRAMP males (n = 5), Tconv cells from the pooled secondary lymphoid organs (SLOs) of untreated wild-type Foxp3GFP males (n = 4), and Treg cells from the SLOs of untreated wild-type Foxp3GFP males (n = 4). Data from the latter two groups and Aire−/− group are taken from ref. 21. Each column represents one biological sample. ND = not detected.

Extended Data Fig. 2 Representative flow cytometric analysis of TCRrg mice expressing Group 1 vs. Group 3 TCRs.

Two Group 1 TCRs (ANT and DAS) and two Group 3 TCRs (SAS and SKV) were expressed as TCRrg mice. >6 weeks after bone marrow reconstitution, TCRrg T cells were directly phenotyped using flow cytometry. a, Representative gating strategy schematic and flow cytometric analysis of Foxp3, CD44, and CD69 expression by splenic Group 1 (top) and Group 3 (bottom) TCRrg T cells (top row, TCRβ+CD4+Thy1.1+) or respective host T cells (TCRβ+CD4+CD45.1+). T cells were selected by gating on TCRβ+B220neg, followed by CD4+CD8βneg, then by Thy1.1+CD45.1neg (for TCRrg T cells) or Thy1.1negCD45.1+ (for host T cells). b, Summary plot of data from a showing the frequency of splenic Group 1 (black symbols) and Group 3 (gray symbols) TCRrg T cells (TCRβ+CD4+Thy1.1+) within TCRβ+CD4+ gate. Summary plot of data from a showing the frequency of B cells (B220+) within CD45.1+ gate. Summary plot of data from a showing the frequency of host CD8α+ cells (CD45.1+CD8α+) within CD45+ gate. Summary plot of data from a showing the frequency of host CD4+ cells (CD45.1+CD4+) within CD45+ gate. Summary plot of data from a showing the frequency of Treg cells (Foxp3+) within host CD4+ gate. Summary plot of data from a showing the percentage of host Tconv cells (CD45.1+CD4+Foxp3neg) expressing CD69. Summary plot of data from a showing the percentage of host Tconv cells (CD45.1+CD4+Foxp3neg) expressing CD44. Summary plot of data from a showing the percentage of host CD8α+ cells (CD45.1+CD8α+) expressing CD69. Summary plot of data from a showing the percentage of host CD8α+ cells (CD45.1+CD8α+) expressing CD44. n = 3–4 per TCRrg. Data are pooled from two independent experiments. Each symbol depicts cells from an individual TCRrg mouse. Bold horizontal lines represent means. Error bars represent means ± SEM. p values were calculated using Student’s t-test (two-tailed) for pooled Group 1 clones versus pooled Group 3 clones. ns, p ≥ 0.05, not significant. Source data contain exact p values and group sizes.

Source data

Extended Data Fig. 3 Multiple recurrent prostate-infiltrating CD4+ Tconv clones exhibit hallmarks of steady-state activation.

Three Group 1 and four Group 3 TCRs were expressed as TCRrg mice. >6 weeks after bone marrow reconstitution, TCRrg T cells were directly phenotyped using flow cytometry. a, Summary plot of data from Fig. 2a, b showing the percentage of CD69 expression by Group 1 (black symbols) and Group 3 (gray symbols) TCRrg T cells (TCRβ+CD4+Thy1.1+) isolated from the indicated regional lymph nodes of primary TCRrg mice (n = 1–7 per TCRrg). Data are pooled from eleven independent experiments. Each symbol depicts cells from an individual TCRrg mouse. iLN, inguinal lymph nodes; aLN, axillary lymph nodes; bLN, brachial lymph nodes; cLN, cervical lymph nodes; pLN, para-aortic lymph nodes; mLN, mesenteric lymph nodes. b, Representative flow cytometric analysis of Ki67 expression by Group 1 (top) and Group 3 (bottom) TCRrg T cells (TCRβ+CD4+Thy1.1+) isolated from the spleen of primary TCRrg mice. SP, spleen. c, Summary plot of data from b showing the percentage of Ki67 expression by Group 1 (black symbols) and Group 3 (gray symbols) TCRrg T cells (TCRβ+CD4+Thy1.1+) isolated from the spleen or pooled lymph nodes of primary TCRrg mice (n = 1–8 per TCRrg). Data are pooled from eight independent experiments. Each symbol depicts cells from an individual TCRrg mouse. SP, spleen; LN, pooled lymph nodes (which include inguinal, axillary, brachial, cervical, para-aortic, and mesenteric lymph nodes). Bold horizontal lines represent means. Error bars represent means ± s.e.m. Two-sided p values were calculated using Student’s t-tests for pooled Group 1 clones versus pooled Group 3 clones. ns, p ≥ 0.05. ns, not significant. Source data contain exact p values and group sizes.

Source data

Extended Data Fig. 4 Recurrent CD4+ Tconv clones detected in the prostate of Treg cell-depleted mice can be binned into three groups based on hallmarks of steady-state activation and reactivity to MHC-II-restricted self-ligands.

Three Group 1, six Group 2, and four Group 3 TCRs as defined in the text and Supplementary Table 2 were cloned and expressed as TCRrg mice. TCRrg T cells were purified and subjected to in vitro reactivity assays. a, Purified TCRβ+CD4+Thy1.1+ TCRrg T cells expressing the indicated Group 1 (top), Group 2 (middle), and Group 3 (bottom) clones were labeled with CellTrace Violet (CTV) and co-cultured with splenic CD11c+ dendritic cells (DCs) and mouse recombinant IL-2 (mrIL-2) for 5 days (see Methods). Representative flow cytometric analyses of CTV dilution are shown. b, Summary plot of data from a depicting the percentage of CTV-diluted Group 1 (black symbols), Group 2 (white symbols), and Group 3 (gray symbols) TCRrg T cells (n = 2–10 per TCRrg). Data are pooled from eight independent experiments. Each symbol represents an individual co-culture. c, Histograms displaying representative flow cytometric analysis of proliferation by Group 1 CTV-labeled TCRrg cells (TCRβ+CD4+Thy1.1+) co-cultured with splenic CD11c+ DCs and mrIL-2 ± protein extracts isolated from prostatic lysates of untreated Foxp3WT or Treg-depleted Foxp3DTR-EGFP males for 5 days (see Methods). Far left column, TCRrg cells co-cultured with mrIL-2 in the absence of CD11c+ DCs served as negative controls. Far right column, TCRrg cells co-cultured with anti-CD3ε/anti-CD28 MACSiBead particles (1:1) and mrIL-2 served as positive controls. Bottom row, purified TCRβ+CD4+ TCRtg T cells expressing the prostate-specific MJ23 transgenic TCR (MJ23tg) served as secondary positive controls, indicated by turquoise-colored histograms. d, Summary plot of data from c depicting the percentage of CTV-diluted Group 1 TCRrg (black symbols) and MJ23tg T cells (turquoise symbols) (n = 2–8 per TCRrg or TCRtg). Data are pooled from three independent experiments. Symbols represent individual co-cultures. In b and d, means ± s.e.m. are indicated. For b, two-sided p values were calculated using Kruskal–Wallis tests and Dunn’s multiple comparison tests, with clones pooled by group. For d, two-sided p values were calculated using Kruskal–Wallis tests and Dunn’s multiple comparison tests separately for each clone, comparing each experimental condition to the ‘No DC’ control. ns, p ≥ 0.05, not significant. Source data contain exact p values and group sizes.

Source data

Extended Data Fig. 5 Group 3 clones express common hallmarks of T follicular helper cells.

As shown in Fig. 3, comparative RNA-Seq analysis of Group 3 SAS and Group 1 ANT TCRrg cells. The SAS and ANT TCRs were cloned and expressed as TCRrg mice (see Methods). TCRrg T cells were purified from the pooled secondary lymphoid organs (SLOs) of primary TCRrg mice and subjected to bulk RNA sequencing (see Methods). Only biological samples containing ≥ 1 × 105 TCRrg T cells were included for analysis (n = 4 mice per TCR). a, Gating strategy schematic for the fluorescence-activated cells sorting (FACS) of TCRrg T cells for RNA sequencing. TCRrg T cells were selected by gating on Dumpneg (B220negCD11bnegCD11cnegF4/80neg), followed by CD4+CD8βneg, then by Thy1.1+CD45.1neg. b, RNA-seq volcano plot depicting differential gene expression for Group 3 SAS and Group 1 ANT TCRrg cells. The –log10 q-value versus log2 fold-change is depicted for genes with average log2 CPM ≥ −1. Blue and red dots denote genes over-represented in Group 1 ANT and Group 3 SAS TCRrg T cells, respectively, with FDR ≤ 0.05. Labels denote top genes that are over- or underrepresented, q ≤ 1 × 10−50 (82 genes). Data are pooled from two independent experiments. q values were generated using edgeR (see Methods). c, Principal component analysis (PCA) of mRNA expression in Group 1 ANT and Group 3 SAS TCRrg T cells, presented as log-scaled normalized expression (log2 CPM). Each dot corresponds to an independent biological sample. Green and magenta dots denote samples isolated from Group 1 ANT and Group 3 SAS TCRrg mice, respectively. n = 4 per TCR. Data are pooled from two independent experiments.

Extended Data Fig. 6 The frequency and phenotype of Group 3 SAS T cells is not impacted following transfer to B cell-depleted recipients.

The Group 1 ANT TCR and Group 3 SAS TCRs were cloned and expressed as TCRrg mice. 1 × 105 FACS-purified TCRrg cells were transferred into treated recipient mice. 7 d prior to TCRrg cell transfer, recipient mice were treated with anti-CD20 or control antibody (see Methods). Donor cell frequency and phenotype was assessed 7 d post-transfer. a, Representative flow cytometric analysis showing the efficiency of B cell depletion in control and anti-CD20 treated recipient mice. SP, spleen; LN, pooled lymph nodes; PP, Peyer’s Patches. b, Summary plot of data from a showing the frequency of B cells (CD19+TCRbneg) recovered from the indicated sites of control- and anti-CD20-treated recipient mice (n = 10–12 per condition). Data pooled from two independent experiments. Each symbol depicts cells from an individual TCRrg mouse. c, Representative flow showing the frequency of CD45.2+CD45.1neg donor TCRrg cells recovered from the spleen of control- and anti-CD20-treated recipient mice. d, Summary plot of data from c showing the frequency of ANTrg and SASrg donor T cells in the spleen of recipient mice treated with control or anti-CD20 antibody (n = 5–7 per condition). Data pooled from two independent experiments. e, Representative flow analysis of donor TCRrg T cells expressing the indicated TCRs in either control or anti-CD20 treated recipients. Histograms of expression of the indicated markers are shown for the indicated TCR and treatment group. The percentage of cells falling within the indicated gates is denoted. f, Summary plots of data from e. For control treated mice and anti-CD20 treated mice, the percentage of cells expressing the indicated marker is plotted for the indicated TCR (n = 5–7 per condition). Data pooled from two independent experiments. Bold horizontal lines represent means. Error bars represent means ± s.e.m. Two-sided p values were calculated using unpaired Student’s t-tests and corrected for multiple comparisons using the Holm-Šídák method. ns, p ≥ 0.05, not significant. Source data contain exact p values and group sizes.

Source data

Extended Data Fig. 7 The Group 3 TCR SKV does not facilitate thymic Treg cell development at low clonal frequencies.

The Group 3 SKV TCR was cloned and expressed as low-frequency TCRrg mixed bone marrow chimeras (see Methods). TCRrg T cells were directly phenotyped using flow cytometry. Thymocytes were gated on CD73neg cells to distinguish newly developed thymocytes from recirculating mature T cells in the thymus59. a, Representative flow cytometric analysis of thymic (top, CD73negTCRβ+Thy1.1+) and splenic (bottom, TCRβ+CD4+Thy1.1+) TCRrg T cell frequency recovered from low-frequency TCRrg mixed bone marrow chimeras. b, Representative flow cytometric analysis of Foxp3 vs. CD4 expression by thymic CD4SP (top, CD73neg TCRβhiCD4+CD8βnegThy1.1+) and splenic CD4+ (bottom, TCRβ+CD4+Thy1.1+) TCRrg T cells from low-frequency TCRrg mixed bone marrow chimeras. Far right column, thymic CD4SP (CD73negTCRβhiCD4+CD8βneg) and splenic CD4+ (TCRβ+CD4+) T cells from untreated B6.SJL mice served as positive controls. c, Representative flow cytometric analysis of Foxp3 vs. CD69 (top) and Ki67 (bottom) expression by splenic CD4+ (TCRβ+CD4+Thy1.1+) TCRrg T cells from low-frequency TCRrg mixed bone marrow chimeras. Far right column, splenic CD4+ (TCRβ+CD4+) T cells from untreated B6.SJL mice served as controls. Data is representative of at least two independent experiments. Representative flow plots are ordered from highest (left) to lowest (right) thymic TCRrg frequency.

Extended Data Fig. 8 Additional phenotyping of TCRrg thymocytes expressing Group 1 vs. Group 3 TCRs.

Two Group 1 TCRs (ANT and DAS) and two Group 3 TCRs (SAS and SKV) were cloned and expressed as TCRrg mice. Thymi from TCRrg mice were isolated and depleted of CD8α-expressing cells prior to phenotyping by flow cytometry. Thymocytes isolated from polyclonal wild-type (WT) mice served as comparative controls. a, Representative flow cytometric analysis of thymocytes expressing the indicated TCRs. Normalized histograms of number of cells vs. expression of the indicated markers are shown for CD4+CD8+ DP cells (top row, gated on CD73negTCRβhiThy1.1+CD4+CD8β+) or CD4SP cells (bottom row, gated on CD73negTCRβhiThy1.1+CD4+CD8βneg). The percentage of cells falling within each indicated gate is denoted. b, Summary plots of data from a. For CD4+CD8+ DP cells (top row) or CD4SP cells (bottom row), the percentage of polyclonal WT (open symbols), Group 1 (black symbols), or Group 3 (gray symbols) expressing the indicated markers is depicted (n = 3–4 per TCRrg, n = 2 per polyclonal WT group). Data are pooled from two independent experiments. Each symbol represents an individual mouse. Bold horizontal lines represent means. Error bars represent means ± s.e.m. Two-sided p values were calculated using Student’s t-tests for pooled Group 1 clones versus pooled Group 3 clones. ns, p ≥ 0.05, not significant. Source data contain exact p values and group sizes.

Source data

Extended Data Fig. 9 Group 3 SAS T cells proliferate and adopt a TFH-like phenotype following intravenous transfer of CD4SP thymocytes.

The Group 1 ANT TCR and Group 3 SAS TCRs were cloned and expressed as TCRrg mice. Thymi from TCRrg mice were isolated and depleted of CD8α-expressing cells. 5 × 106 SASrg thymocytes or 5 × 105 ANTrg thymocytes were transferred intravenously into B6.SJL recipient mice and analyzed at day 4 or day 11 post-transfer via flow cytometry. a, Representative analysis of CD45.2+CD45.1neg donor TCRrg cells recovered from the spleens of recipient mice at the indicated time points. The percentage of cells falling within the indicated gates are denoted. b, Summary plots of data shown in a (n = 6 per condition). Data are pooled from two independent experiments. Each symbol represents an individual mouse. c, Representative flow cytometric analysis of donor T cells (TCRβ+CD4+CD45.2+Thy1.1+) expressing the indicated TCRs. Normalized histograms of number of cells vs. expression of the indicated markers are shown for the donor cell thymocyte suspension (‘input thymocytes’, top row), cells recovered from the spleen of recipients at day 4 (middle row), and cells recovered from the spleen of recipients at day 11 (bottom row). The percentage of cells falling within the indicated gates is denoted. Splenocytes isolated from polyclonal wild-type (WT) mice served as comparative controls. d, Summary plots of data from c. For donor ANTrg T cells (black symbols) and donor SASrg T cells (gray symbols), the percentage of cells expressing the indicated markers is plotted for the indicated time points (n = 2–6 per condition). Data are pooled from two independent experiments. Each symbol represents an individual mouse. Bold horizontal lines represent means. Error bars represent means ± s.e.m. Two-sided p values were calculated using Student’s t-tests and corrected for multiple comparisons using the Holm-Šídák method. ns, p ≥ 0.05, not significant. Source data contain exact p values and group sizes.

Source data

Extended Data Table 1 Gene set enrichment analysis of Group 1 and Group 3 TCRrg Tconv cells

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Table 1 Complete TCRα sequencing frequency table. For samples subjected to complete TCRα sequencing (Methods), the frequencies of each TCR CDR3α segment, as a percentage of all TCRα sequence reads within a given sample, are listed. For this table, TCRs were catalogued solely based on predicted TCR CDR3α sequence, regardless of TRAV usage. The table lists the Sample IDs (PR 1–5, SG 1–5 and TRAMP 1–5) for the complete TCRα sequencing dataset, the organ from which T cells were isolated (PR and SGs), the genotype (Foxp3DTR-EGFP or TRAMP), the treatment (Treg-depleted or untreated), and the T cell subset (Tconv cells). For each biological sample, the total number of in-frame TCR sequence reads and the number of unique TCR sequence reads are also listed. Supplementary Table 2 Selection of recurrent prostate-infiltrating CD4+ Tconv clones for TCR retrogenic studies. Thirteen of the top 20 most abundant recurrent TCRα sequences expressed by Tconv cells in the prostate of Treg cell-depleted mice were selected for TCR retrogenic studies. TCRs were selected based on differential TRAV and TRAJ usage. Each TCRα chain is denoted using a three-letter code representing residues 3–5 of the predicted CDR3α chain. The full TCRα sequence information is listed, including TRAV and TRAJ usage. The 13 selected TCRα clonotypes can be binned into three groups based on hallmarks of steady-state activation and reactivity to MHC-II-restricted self-ligands (Extended Data Figs. 3 and 4). Supplementary Table 3 List of primers used for the generation of TCR retrogenic constructs.

Source data

Source Data Fig. 1

Statistical source data and exact P values.

Source Data Fig. 2

Statistical source data and exact P values.

Source Data Fig. 3

Statistical source data and exact P values.

Source Data Fig. 4

Statistical source data and exact P values.

Source Data Fig. 5

Statistical source data and exact P values.

Source Data Fig. 6

Statistical source data and exact P values.

Source Data Extended Data Fig. 2

Statistical source data and exact P values.

Source Data Extended Data Fig. 3

Statistical source data and exact P values.

Source Data Extended Data Fig. 4

Statistical source data and exact P values.

Source Data Extended Data Fig. 6

Statistical source data and exact P values.

Source Data Extended Data Fig. 8

Statistical source data and exact P values.

Source Data Extended Data Fig. 9

Statistical source data and exact P values.

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

Lee, V., Rodriguez, D.M., Ganci, N.K. et al. The endogenous repertoire harbors self-reactive CD4+ T cell clones that adopt a follicular helper T cell-like phenotype at steady state. Nat Immunol 24, 487–500 (2023). https://doi.org/10.1038/s41590-023-01425-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01425-0

This article is cited by

Search

Quick links

Nature Briefing

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

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