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:

Regulatory T cells function in established systemic inflammation and reverse fatal autoimmunity

Abstract

The immunosuppressive function of regulatory T (Treg) cells is dependent on continuous expression of the transcription factor Foxp3. Foxp3 loss of function or induced ablation of Treg cells results in a fatal autoimmune disease featuring all known types of inflammatory responses with every manifestation stemming from Treg cell paucity, highlighting a vital function of Treg cells in preventing fatal autoimmune inflammation. However, a major question remains whether Treg cells can persist and effectively exert their function in a disease state, where a broad spectrum of inflammatory mediators can either inactivate Treg cells or render innate and adaptive pro-inflammatory effector cells insensitive to suppression. By reinstating Foxp3 protein expression and suppressor function in cells expressing a reversible Foxp3 null allele in severely diseased mice, we found that the resulting single pool of rescued Treg cells normalized immune activation, quelled severe tissue inflammation, reversed fatal autoimmune disease and provided long-term protection against them. Thus, Treg cells are functional in settings of established broad-spectrum systemic inflammation and are capable of affording sustained reset of immune homeostasis.

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: Restoration of Foxp3 expression in Treg cell ‘wannabes’ cures fulminant autoimmunity in male Foxp3LSL mice.
Fig. 2: Restoration of Foxp3 expression in Treg cell ‘wannabes’ in mosaic adult female Foxp3LSL/DTR–GFP mice suppresses immune activation caused by DT-mediated Treg cell ablation.
Fig. 3: Rescued Treg cells in inflamed mice are activated and potently suppressive in inflammatory settings.
Fig. 4: Rescued Treg cells in male Foxp3LSL mice provide long-term protection from autoimmune inflammatory disease.
Fig. 5: Single-cell transcriptomic analysis of control and long-lived rescued Treg cells.
Fig. 6: Identification and analysis of γREG+ Treg cells from the single-cell RNA-sequencing data.

Similar content being viewed by others

Data availability

All sequencing data generated in this study can be accessed at the Gene Expression Omnibus (GEO) under accession GSE179710. The custom mouse genome used for scRNA-seq analysis, which was generated by adding the tdTomato sequence to GRCm38 (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.20/), is also available at the GEO. The following gene-set enrichment analysis gene sets were used for data analysis: GO_CELL_CYCLE_G1_S_PHASE_TRANSITION, GO_CELL_CYCLE_G2_M_PHASE_TRANSITION, GO_DNA_REPLICATION, HALLMARK_APOPTOSIS, HALLMARK_FATTY_ACID_METABOLISM, HALLMARK_G2M_CHECKPOINT, HALLMARK_GLYCOLYSIS, HALLMARK_IL2_STAT5_SIGNALING, HALLMARK_OXIDATIVE_PHOSPHORYLATION, HALLMARK_PI3K_AKT_MTOR_SIGNALING, HALLMARK_REACTIVE_OXYGEN_SPECIES_PATHWAY, HALLMARK_TGF_BETA_SIGNALING, HALLMARK_WNT_BETA_CATENIN_SIGNALING, KEGG_CITRATE_CYCLE_TCA_CYCLE, KEGG_PURINE_METABOLISM and REACTOME_EUKARYOTIC_TRANSLATION_INITIATION. Source data are provided with this paper.

Code availability

All custom scripts are available upon request to the corresponding authors.

References

  1. Kanangat, S. et al. Disease in the scurfy (sf) mouse is associated with overexpression of cytokine genes. Eur. J. Immunol. 26, 161–165 (1996).

    Article  CAS  PubMed  Google Scholar 

  2. Chatila, T. A. Role of regulatory T cells in human diseases. J. Allergy Clin. Immunol. 116, 949–959 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Godfrey, V. L., Wilkinson, J. E. & Russell, L. B. X-linked lymphoreticular disease in the scurfy (sf) mutant mouse. Am. J. Pathol. 138, 1379–1387 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Khattri, R., Cox, T., Yasayko, S. A. & Ramsdell, F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat. Immunol. 4, 337–342 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Fontenot, J. D., Gavin, M. A. & Rudensky, A. Y. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat. Immunol. 4, 330–336 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. Fontenot, J. D. et al. Regulatory T cell lineage specification by the forkhead transcription factor Foxp3. Immunity 22, 329–341 (2005).

    Article  CAS  PubMed  Google Scholar 

  7. Hori, S., Nomura, T. & Sakaguchi, S. Control of regulatory T cell development by the transcription factor Foxp3. Science 299, 1057–1061 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Gavin, M. A. et al. Foxp3-dependent programme of regulatory T cell differentiation. Nature 445, 771–775 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Lin, W. et al. Regulatory T cell development in the absence of functional Foxp3. Nat. Immunol. 8, 359–368 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Ohkura, N. et al. T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity 37, 785–799 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Brunkow, M. E. et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat. Genet. 27, 68–73 (2001).

    Article  CAS  PubMed  Google Scholar 

  12. Ramsdell, F. & Ziegler, S. F. FOXP3 and scurfy: how it all began. Nat. Rev. Immunol. 14, 343–349 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. 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 

  14. Gangaplara, A. et al. Type I interferon signaling attenuates regulatory T cell function in viral infection and in the tumor microenvironment. PLoS Pathog. 14, e1006985 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Izcue, A. et al. Interleukin-23 restrains regulatory T cell activity to drive T cell-dependent colitis. Immunity 28, 559–570 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Nish, S. A. et al. T cell-intrinsic role of IL-6 signaling in primary and memory responses. Elife 3, e01949 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Overacre-Delgoffe, A. E. et al. Interferon-gamma drives Treg fragility to promote anti-tumor immunity. Cell 169, 1130–1141 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pasare, C. & Medzhitov, R. Toll pathway-dependent blockade of CD4+CD25+ T cell-mediated suppression by dendritic cells. Science 299, 1033–1036 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Pelly, V. S. et al. Interleukin 4 promotes the development of ex-Foxp3 TH2 cells during immunity to intestinal helminths. J. Exp. Med 214, 1809–1826 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Srivastava, S., Koch, M. A., Pepper, M. & Campbell, D. J. Type I interferons directly inhibit regulatory T cells to allow optimal antiviral T cell responses during acute LCMV infection. J. Exp. Med 211, 961–974 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Korn, T. et al. Myelin-specific regulatory T cells accumulate in the CNS but fail to control autoimmune inflammation. Nat. Med 13, 423–431 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Clough, L. E. et al. Release from regulatory T cell-mediated suppression during the onset of tissue-specific autoimmunity is associated with elevated IL-21. J. Immunol. 180, 5393–5401 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Gao, Y. et al. Inflammation negatively regulates FOXP3 and regulatory T cell function via DBC1. Proc. Natl Acad. Sci. USA 112, E3246–E3254 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Komatsu, N. et al. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat. Med. 20, 62–68 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Bailey-Bucktrout, S. L. et al. Self-antigen-driven activation induces instability of regulatory T cells during an inflammatory autoimmune response. Immunity 39, 949–962 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Tian, L. et al. Foxp3+ regulatory T cells exert asymmetric control over murine helper responses by inducing TH2 cell apoptosis. Blood 118, 1845–1853 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Scherer, M. T., Ignatowicz, L., Winslow, G. M., Kappler, J. W. & Marrack, P. Superantigens: bacterial and viral proteins that manipulate the immune system. Annu. Rev. Cell Biol. 9, 101–128 (1993).

    Article  CAS  PubMed  Google Scholar 

  28. Pacholczyk, R., Kraj, P. & Ignatowicz, L. Peptide specificity of thymic selection of CD4+CD25+ T cells. J. Immunol. 168, 613–620 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Romagnoli, P., Hudrisier, D. & van Meerwijk, J. P. Preferential recognition of self antigens despite normal thymic deletion of CD4+CD25+ regulatory T cells. J. Immunol. 168, 1644–1648 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Arvey, A. et al. Inflammation-induced repression of chromatin bound by the transcription factor Foxp3 in regulatory T cells. Nat. Immunol. 15, 580–587 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chinen, T. et al. An essential role for the IL-2 receptor in Treg cell function. Nat. Immunol. 17, 1322–1333 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Vignali, D. A., Collison, L. W. & Workman, C. J. How regulatory T cells work. Nat. Rev. Immunol. 8, 523–532 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Shevach, E. M. Mechanisms of Foxp3+ T regulatory cell-mediated suppression. Immunity 30, 636–645 (2009).

    Article  CAS  PubMed  Google Scholar 

  34. 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 

  35. Levine, A. G. et al. Suppression of lethal autoimmunity by regulatory T cells with a single TCR specificity. J. Exp. Med. 214, 609–622 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Feng, Y. et al. A mechanism for expansion of regulatory T cell repertoire and its role in self-tolerance. Nature 528, 132–136 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ye, J. et al. The aryl hydrocarbon receptor preferentially marks and promotes gut regulatory T cells. Cell Rep. 21, 2277–2290 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Pritykin, Y. et al. A unified atlas of CD8 T cell dysfunctional states in cancer and infection. Mol. Cell 81, 2477–2493 (2021).

    Article  CAS  PubMed  Google Scholar 

  40. Pierson, W. et al. Antiapoptotic Mcl-1 is critical for the survival and niche-filling capacity of Foxp3+ regulatory T cells. Nat. Immunol. 14, 959–965 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Setty, M. et al. Characterization of cell fate probabilities in single-cell data with Palantir. Nat. Biotechnol. 37, 451–460 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Miragaia, R. J. et al. Single-cell transcriptomics of regulatory T cells reveals trajectories of tissue adaptation. Immunity 50, 493–504 e497 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Fontenot, J. D., Rasmussen, J. P., Gavin, M. A. & Rudensky, A. Y. A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat. Immunol. 6, 1142–1151 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Xing, S. et al. Tcf1 and Lef1 are required for the immunosuppressive function of regulatory T cells. J. Exp. Med. 216, 847–866 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yang, B. H. et al. TCF1 and LEF1 control Treg competitive survival and Tfr development to prevent autoimmune diseases. Cell Rep. 27, 3629–3645 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Allenspach, E. & Torgerson, T. R. Autoimmunity and primary immunodeficiency disorders. J. Clin. Immunol. 1, 57–67 (2016).

    Article  CAS  Google Scholar 

  47. Georgiev, P., Charbonnier, L. M. & Chatila, T. A. Regulatory T cells: the many faces of Foxp3. J. Clin. Immunol. 39, 623–640 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Park, J. H. et al. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome: a systematic review. Autoimmun. Rev. 19, 102526 (2020).

    Article  CAS  PubMed  Google Scholar 

  49. Oldenhove, G. et al. Decrease of Foxp3+ Treg cell number and acquisition of effector cell phenotype during lethal infection. Immunity 31, 772–786 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Schenten, D. et al. Signaling through the adaptor molecule MyD88 in CD4+ T cells is required to overcome suppression by regulatory T cells. Immunity 40, 78–90 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lund, J. M., Hsing, L., Pham, T. T. & Rudensky, A. Y. Coordination of early protective immunity to viral infection by regulatory T cells. Science 320, 1220–1224 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ruckwardt, T. J., Bonaparte, K. L., Nason, M. C. & Graham, B. S. Regulatory T cells promote early influx of CD8+ T cells in the lungs of respiratory syncytial virus-infected mice and diminish immunodominance disparities. J. Virol. 83, 3019–3028 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hall, A. O. et al. The cytokines interleukin 27 and interferon-gamma promote distinct Treg cell populations required to limit infection-induced pathology. Immunity 37, 511–523 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Durant, L. R. et al. Regulatory T cells prevent TH2 immune responses and pulmonary eosinophilia during respiratory syncytial virus infection in mice. J. Virol. 87, 10946–10954 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Schmitz, I. et al. IL-21 restricts virus-driven Treg cell expansion in chronic LCMV infection. PLoS Pathog. 9, e1003362 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zhao, J., Zhao, J. & Perlman, S. Virus-specific regulatory T cells ameliorate encephalitis by repressing effector T cell functions from priming to effector stages. PLoS Pathog. 10, e1004279 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Long, S. A. & Buckner, J. H. CD4+FOXP3+ T regulatory cells in human autoimmunity: more than a numbers game. J. Immunol. 187, 2061–2066 (2011).

    Article  CAS  PubMed  Google Scholar 

  58. Venken, K. et al. Natural naive CD4+CD25+CD127lo regulatory T cell development and function are disturbed in multiple sclerosis patients: recovery of memory Treg homeostasis during disease progression. J. Immunol. 180, 6411–6420 (2008).

    Article  CAS  PubMed  Google Scholar 

  59. Ferraro, A. et al. Interindividual variation in human T regulatory cells. Proc. Natl Acad. Sci. USA 111, E1111–E1120 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Arvey, A. et al. Genetic and epigenetic variation in the lineage specification of regulatory T cells. Elife 4, e07571 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Nasrallah, R. et al. A distal enhancer at risk locus 11q13.5 promotes suppression of colitis by Treg cells. Nature 583, 447–452 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mottet, C., Uhlig, H. H. & Powrie, F. Cutting edge: cure of colitis by CD4+CD25+ regulatory T cells. J. Immunol. 170, 3939–3943 (2003).

    Article  CAS  PubMed  Google Scholar 

  63. Uhlig, H. H. et al. Characterization of Foxp3+CD4+CD25+ and IL-10-secreting CD4+CD25+ T cells during cure of colitis. J. Immunol. 177, 5852–5860 (2006).

    Article  CAS  PubMed  Google Scholar 

  64. Wan, Y. Y. & Flavell, R. A. Regulatory T cell functions are subverted and converted owing to attenuated Foxp3 expression. Nature 445, 766–770 (2007).

    Article  CAS  PubMed  Google Scholar 

  65. Josefowicz, S. Z., Lu, L. F. & Rudensky, A. Y. Regulatory T cells: mechanisms of differentiation and function. Annu Rev. Immunol. 30, 531–564 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Sakaguchi, S. et al. Regulatory T cells and human disease. Annu. Rev. Immunol. 38, 541–566 (2020).

    Article  CAS  PubMed  Google Scholar 

  67. Tahvildari, M. & Dana, R. Low-dose IL-2 therapy in transplantation, autoimmunity and inflammatory diseases. J. Immunol. 203, 2749–2755 (2019).

    Article  CAS  PubMed  Google Scholar 

  68. Vogtenhuber, C. et al. Constitutively active Stat5b in CD4+ T cells inhibits graft-versus-host disease lethality associated with increased regulatory T cell potency and decreased T effector cell responses. Blood 116, 466–474 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Sledzinska, A. et al. TGF-beta signalling is required for CD4+ T cell homeostasis but dispensable for regulatory T cell function. PLoS Biol. 11, e1001674 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Mombaerts, P. et al. Mutations in T cell antigen receptor genes alpha and beta block thymocyte development at different stages. Nature 360, 225–231 (1992).

    Article  CAS  PubMed  Google Scholar 

  71. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  72. Campbell, C. et al. Extrathymically generated regulatory T cells establish a niche for intestinal border-dwelling bacteria and affect physiologic metabolite balance. Immunity 48, 1245–1257 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  74. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 20, 1297–1303 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  76. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Levine, J. H. et al. Data-driven phenotypic dissection of AML reveals progenitor-like cells that correlate with prognosis. Cell 162, 184–197 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Bolotin, D. A. et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380–381 (2015).

    Article  CAS  PubMed  Google Scholar 

  81. Shugay, M. et al. VDJtools: unifying post-analysis of T cell receptor repertoires. PLoS Comput Biol. 11, e1004503 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank E. Azizi for advice on scRNA-seq data analysis and members of the laboratory of A.Y.R. for technical support and discussion, Memorial Sloan Kettering Cancer Center (MSKCC) Single Cell Research Initiative and Integrated Genomics Operation Core facility for single-cell and bulk RNA-seq. This work was supported by the National Institutes of Health (grant R01 AI034206 to A.Y.R.), an Irvington-Cancer Research Institute Postdoctoral Fellowship (to W.H.), the Ludwig Center at Memorial Sloan Kettering, Parker Institute for Cancer Immunotherapy (PICI) (to A.Y.R.) and the National Cancer Institute Cancer Center (grant P30 CA08748). A.Y.R is an investigator with the Howard Hughes Medical Institute. Z.-M.W. is supported by a Bruce Charles Forbes Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

W.H. and A.Y.R. conceived the study and designed experiments. W.H., Z.-M.W. and A.Y.R. interpreted the data and wrote the manuscript. W.H. and Z.-M.W. performed the experiments. Y.F. designed the targeting construct and generated the Foxp3LSL mouse. B.E.H. performed the antibody ELISA assays. J.G.V., J.v.d.V. and R.B.-P. provided assistance with some experiments. J.G.V. assisted with mouse generation and animal colony maintenance. Z.-M.W. and M.S. performed analysis of the bulk RNA-seq experiment. M.S. performed analysis of scRNA-seq and TCR-sequencing datasets. W.H. and A.Y.R. supervised the study. Correspondence and requests for materials should be addressed to the corresponding authors.

Corresponding authors

Correspondence to Wei Hu or Alexander Y. Rudensky.

Ethics declarations

Competing interests

A.Y.R. is a co-founder and Science Advisory Board member of and holds stock options in Vedanta Bioscience and Sonoma Biotherapeutics. The other authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Masahiro Ono and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Generation and characterization of Foxp3LSL mice.

a, Schematics of the targeting construct and the Foxp3LSL allele before and after Flp recombinase-mediated removal of the neo cassette. Homologous regions between the targeting construct and the WT allele are demarcated with dashed gray lines. Gene map is based on RefSeq record NM_001199347.1. pA, bovine growth hormone polyadenylation signal; neo, neomycin resistance gene; PGK, mouse phosphoglycerate kinase 1 promoter; 3xSV40pA, triple-tandem SV40 early polyadenylation signals (STOP cassette); DTA, diphtheria toxin A. b, Genotyping PCR showing the WT (275 bp) and knock-in-specific (350 bp) bands using primers labeled in a. Gel picture is reproduced for and representative of all animals analyzed in this study. c, d, Flow cytometric analysis of T cells and TCRβhi single positive thymocytes from 3-week-old male Foxp3LSL/y (c) and 8–10-week-old female Foxp3LSL/WT (d) mice. LN, lymph nodes. e, Expression of molecules associated with T cell activation in splenic Foxp3+ Treg and Thy1.1+ Treg ‘wannabes’, and naïve (CD44loCD62Lhi) and activated (CD44hiCD62Llo) conventional CD4 T cells shown in d. f, Percentages of TCRVβ5+, Vβ6+ and Vβ8+ cells among the indicated splenic CD4 T cell populations from 8–10-week-old female Foxp3LSL/DTR–GFP mice. Cells are gated on the CD44loCD62Lhi subset to avoid potential clonal expansion during activation. One-way ANOVA. g, 2×106 CD45.1+Foxp3 conventional CD4 T cells were co-transferred with 2×105 CD45.2+ Treg ‘wannabes’ from 2–3-week-old Foxp3LSL/y mice or Treg cells from 6–8-week-old Foxp3GFP mice into Tcrb–/–Tcrd–/– mice. Recipients were orally gavaged with tamoxifen and injected i.p. with diphtheria toxin (DT) at the indicated time points (top). DT was administered to deplete the few contaminating DTR-expressing Treg cells in the transferred FACS-purified conventional T cells or those induced to express Foxp3 after transfer. Cellularity and proliferation of CD45.1+ responder cells from lymph nodes were analyzed using flow cytometry (bottom). Data are representative of two independent experiments. All error bars denote mean ± s.e.m. ns, non-significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001.

Source data

Extended Data Fig. 2 Restoration of Foxp3 expression reverses spontaneous lymphocyte expansion, myelo-proliferation, and cytokine production, and normalizes circulatory Ig levels in male Foxp3LSL mice.

a, 4-OHT mediated recombination efficiency in lymphoid and non-lymphoid organs. Experimental scheme (left) and recombination efficiency of Rosa26Tom in Treg cells from indicated organs (right). One-way ANOVA. LN, lymph nodes. b-g, Male Foxp3LSL mice were treated with 4-OHT on postnatal day 14 and analyzed throughout the following 4 weeks. b, Representative histograms showing expression of proliferation and activation markers by splenic conventional CD4 and CD8 T cells at the indicated time points. c, Representative contour plots of flow cytometric analyses of activated and cytokine-producing splenic conventional CD4 and CD8 T cell populations. d, e, frequencies (d) and numbers (e) of activated, proliferating and cytokine-producing conventional CD4 and CD8 T cells from lymph nodes of mice of indicated genotypes at designated time points after 4-OHT treatment. Two-way ANOVA with Tukey’s multiple comparison correction. f, Numbers of splenic myeloid cell populations at indicated time points after 4-OHT treatment. Two-way ANOVA with Tukey’s multiple comparison correction. g, Serum antibody levels as in Fig. 1g. One-way ANOVA with Tukey’s multiple comparison test. All error bars denote mean ± s.e.m. ns, non-significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001.

Source data

Extended Data Fig. 3 Restoration of Foxp3 expression in peripheral Treg ‘wannabes’ rescues immune activation in male Foxp3LSL mice.

a, Experimental design. 2-week-old mice were treated with 4-hydroxytamoxifen (4-OHT) while being continuously treated with FTY720 to block thymic output. A Rosa26loxP−STOP−loxP−tdTomato (R26LSL−tdTomato) recombination reporter allele was introduced to the mice to enable labeling of all CD4 T cells undergoing Cre-mediated recombination induced by 4-OHT. b, Number of Treg cells in the thymus (left), and percentage of tdTomato+ cells among Foxp3Thy1.1 CD4SP thymocytes (right), from FTY720-treated mice and untreated controls on day 14. Two-tailed unpaired t-tests with multiple hypothesis testing correction using the Holm-Sidak method. c, d, Numbers of activated, proliferating, and cytokine-producing conventional CD4 and CD8 T cells in the spleen (c) and lymph nodes (d). One-way ANOVA with Tukey’s multiple comparison test. All error bars denote mean ± s.e.m. ns, non-significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001.

Source data

Extended Data Fig. 4 Restoration of Foxp3 expression in Treg ‘wannabes’ rescues tissue damage in the skin and liver of male Foxp3LSL mice.

Mice were treated with 4-OHT on postnatal day 14 and examined at the indicated time points post-treatment. a, b, H&E staining of skin sections showing epidermal hyperplasia and formation of serocellular crust (arrow heads). Dashed lines demarcate the boundary between epidermis and dermis. Images are representative of 4 Foxp3LSL/yCd4wt, 4 Foxp3LSL/yCd4creERT2 and 2 Foxp3DTR−GFP/yCd4creERT2 mice. c, TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) assay followed by immunohistochemistry to visualize apoptotic cells (arrow heads) in H&E counter-stained liver sections from mice of indicated genotypes. d, Quantification of TUNEL+ cells. e, Measurement of serum albumin levels. Two-way ANOVA with Tukey’s multiple comparison. All error bars denote mean ± s.e.m. ns, non-significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001.

Source data

Extended Data Fig. 5 Restoration of Foxp3 expression in Treg ‘wannabes’ in mosaic adult female Foxp3LSL/DTR−GFP mice suppresses immune activation caused by diphtheria toxin-mediated Treg cell ablation.

Experimental scheme shown in Fig. 2a. a, Representative histograms showing expression of activation and proliferation markers in splenic conventional T cell populations. b, Representative contour plots of splenic conventional CD4 and CD8 T cells showing cytokine production. c, Percentages (left) and numbers (right) of Treg and Treg ‘wannabes’ from indicated tissues 5 weeks post 4-OHT administration. Two-way ANOVA with Tukey’s multiple comparison test. All error bars denote mean ± s.e.m. ns, non-significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001. pLN, peripheral (brachial, axillary, and inguinal) lymph nodes; mLN, mesenteric lymph nodes; LP, lamina propria.

Source data

Extended Data Fig. 6 Analysis of gene expression changes in Treg ‘wannabes’ induced upon activation.

a, Experimental scheme. 8–10-week-old heterozygous female Foxp3LSL/DTR−GFPCd4wt mice were treated with diphtheria toxin (DT) on designated days to deplete Foxp3DTR−GFP-expressing Treg cells and induce activation of Foxp3LSL-expressing Treg ‘wannabes’ which were sorted and analyzed by RNA-seq. b, FC-FC plot of activation-induced gene expression changes in Treg cells and ‘wannabes’. Genes with mean normalized counts of >100 are shown. Differentially expressed genes (p < 0.05) are colored based on the direction of the change in either or both cell types. c, Venn diagram showing the numbers of genes with larger than a 2-fold change in activated Treg cells and ‘wannabes’. d, eCDF plots showing expression changes in activated Treg ‘wannabes’ for all genes (black) and Treg activation signature genes that are up- (red) or down- (blue) regulated. Two-sided Kolmogorov-Smirnov test. e, FC-FC plots showing gene expression changes in Treg cells vs. ‘wannabes’ isolated from sick and healthy mice. Signature genes that are up- or down-regulated in activated Treg and conventional CD4 T cells are highlighted in different colors.

Extended Data Fig. 7 Rescued Treg cells in male Foxp3LSL mice exert long-term control of adaptive and innate immune cells.

Mice were treated with a single dose of 4-OHT at 2 weeks of age and analyzed 4 months later. a, Body weight of rescued Foxp3LSLCd4creERT2 and control Foxp3DTR−GFPCd4creERT2 mice over a 4-month time course. Two-way ANOVA with Sidak’s multiple comparison test. b, Analysis of 4-OHT ‘functional’ pharmacokinetics in 2-week-old male mice. Mice were injected with 4-OHT 3–72 hours before transfer of congenically marked recombination-proficient CD4 T cells from Cd4creERT2R26Tom mice. 4-OHT-induced recombination was assayed 7 days later by tdTomato expression among donor CD4 T cells as a readout for 4-OHT activity. Data are pooled from two independent experiments with 2 to 4 mice per time point each. Ctrl, no 4-OHT injection. c, Suppression of in vitro proliferation of conventional CD4 T cells induced by α-CD3 antibody and antigen-presenting cells by control Treg cells from Foxp3DTR−GFP/y or rescued Treg cells from Foxp3LSL/y mice. Two-way ANOVA with Tukey’s multiple comparison test. d-f, Numbers of activated, proliferating, and cytokine-producing conventional CD4 T cells (d), CD8 T cells (e) and myeloid populations (f) in indicated tissues. pLN, peripheral (brachial, axillary, and inguinal) lymph nodes; mLN, mesenteric lymph nodes; LP, lamina propria. Data are pooled from two independent experiments. Two-tailed t-tests with multiple hypothesis testing correction using the Holm-Sidak method. All error bars denote mean ± s.e.m. ns, non-significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001.

Source data

Extended Data Fig. 8 Rescued Treg cell population persisting for 7 months in male Foxp3LSL mice prevents relapse of rampant autoimmunity.

Mice were treated with 4-OHT on postnatal day 14 and analyzed 7 months later. a-c, Frequencies of Treg cells (a) and proliferating, activated, and cytokine-producing conventional CD4 (b) and CD8 (c) T cells. Two-tailed unpaired t-tests with multiple hypothesis testing correction using the Holm-Sidak method. pLN, peripheral (brachial, axillary, and inguinal) lymph nodes; mLN, mesenteric lymph nodes; LP, lamina propria. d, Serum antibody levels. Scales for IgM, IgG1, and IgE were kept the same as in Fig. 1g. Two-tailed unpaired t-test. e, Representative images of hematoxylin and eosin-stained sections of the indicated organs. Images are representative of 5 Foxp3DTR−GFP/y and 5 Foxp3LSL/y mice. f, Clonal diversity of the TCRα repertoire of the long-lived rescued Treg cells from Foxp3LSL/yCd4creERT2 mice at indicated time points after restoring Foxp3 expression upon 4-OHT treatment. The inverse Simpson Index was calculated based on the clone size distribution using clonotypes defined by full nucleotide sequence (left) or CDR3 amino acid sequence (right). g, Total number of unique clones (left) and Gini coefficient (right) of the TCRα repertoire of the long-lived rescued Treg cells. Clonotypes were defined by using the full nucleotide sequence. One-way ANOVA with Dunnett’s multiple hypothesis test. All error bars denote mean ± s.e.m. ns, non-significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001.

Source data

Extended Data Fig. 9 Analysis of long-lived Treg cells in Foxp3LSL and control Foxp3DTR−GFP mice.

Mice were treated with 4-OHT on postnatal day 14 and analyzed 7 months later. a, UMAP visualization of the single-cell transcriptomes colored by imputed expression levels of representative genes. b, Frequencies of CD62Lhi and CD103+ cells among Treg cells in rescued Foxp3LSLCd4creERT2 and control Foxp3DTR−GFPCd4creERT2 mice. Two-tailed multiple t-tests with Holm-Sidak’s correction. pLN, peripheral (brachial, axillary, and inguinal) lymph nodes; mLN, mesenteric lymph nodes; LP, lamina propria. c, Foxp3WTCd4creERT2R26Tom mice were treated with 4-OHT on postnatal day 14 and analyzed 4 months later. Representative contour plots show expression of activation markers by tdTomato+ and tdTomato Foxp3+ CD4 T cells. d, Histogram depicting the density of Foxp3LSL and tdTomato+ or tdTomato Foxp3DTR−GFP Treg cells along the average expression values for the indicated gene sets. All error bars denote mean ± s.e.m. ns, non-significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001.

Source data

Extended Data Fig. 10 Flow cytometric analysis of γREG+ Treg cells.

a, Gating strategy for γREG+ Treg cells in adult mice. b, c, Representative histogram (b) and quantification (c) of IL-4Rα expression in γREG+ and other Treg cell populations identified in a. Two-way ANOVA with Tukey’s multiple comparison correction. d, e, Representative histogram (d) and quantification (e) of CD25 expression in γREG+ and other Treg cell populations identified in a. Two-way ANOVA with Tukey’s multiple comparison correction. f, Percentages of γREG+ Treg cells in lymphoid organs and non-lymphoid tissues of CD45 i.v.-labeled 8–10-week-old Foxp3GFP mice. One-way ANOVA with Sidak’s multiple comparison test. g, Percentages (left) and numbers (right) of γREG+ Treg cells in different organs of 2- or 8-week-old Foxp3GFP−BirA−AVI−TEV mice. Two-sided unpaired t-tests with correction for multiple hypothesis testing using the Holm-Sidak method. All error bars denote mean ± s.e.m. ns, non-significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001. pLN, peripheral (brachial, axillary, and inguinal) lymph nodes; mLN, mesenteric lymph nodes; cLP, colonic lamina propria.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1

Reporting Summary

Supplementary Table 1

Differentially expressed genes for clusters identified in the scRNA-seq analysis as shown in Fig. 6d.

Supplementary Table 2

Sample sizes for all summary data.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed DNA gel picture.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hu, W., Wang, ZM., Feng, Y. et al. Regulatory T cells function in established systemic inflammation and reverse fatal autoimmunity. Nat Immunol 22, 1163–1174 (2021). https://doi.org/10.1038/s41590-021-01001-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01001-4

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