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:

CTLA-4-expressing ILC3s restrain interleukin-23-mediated inflammation

Abstract

Interleukin (IL-)23 is a major mediator and therapeutic target in chronic inflammatory diseases that also elicits tissue protection in the intestine at homeostasis or following acute infection1,2,3,4. However, the mechanisms that shape these beneficial versus pathological outcomes remain poorly understood. To address this gap in knowledge, we performed single-cell RNA sequencing on all IL-23 receptor-expressing cells in the intestine and their acute response to IL-23, revealing a dominance of T cells and group 3 innate lymphoid cells (ILC3s). Unexpectedly, we identified potent upregulation of the immunoregulatory checkpoint molecule cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) on ILC3s. This pathway was activated by gut microbes and IL-23 in a FOXO1- and STAT3-dependent manner. Mice lacking CTLA-4 on ILC3s exhibited reduced regulatory T cells, elevated inflammatory T cells and more-severe intestinal inflammation. IL-23 induction of CTLA-4+ ILC3s was necessary and sufficient to reduce co-stimulatory molecules and increase PD-L1 bioavailability on intestinal myeloid cells. Finally, human ILC3s upregulated CTLA-4 in response to IL-23 or gut inflammation and correlated with immunoregulation in inflammatory bowel disease. These results reveal ILC3-intrinsic CTLA-4 as an essential checkpoint that restrains the pathological outcomes of IL-23, suggesting that disruption of these lymphocytes, which occurs in inflammatory bowel disease5,6,7, contributes to chronic inflammation.

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 single-cell atlas of IL-23 responses in the small intestine identifies CTLA-4+ ILC3s.
Fig. 2: Microbial exposure elicits IL-23 to upregulate CTLA-4 on ILC3s.
Fig. 3: CTLA-4+ ILC3s restrain IL-23-dependent gut inflammation.
Fig. 4: CTLA-4+ ILC3s regulate co-stimulatory and inhibitory checkpoints on myeloid cells in the gut.
Fig. 5: IL-23 upregulates CTLA-4+ ILC3s in humans and this axis is altered in IBD.

Similar content being viewed by others

Data availability

All data necessary to understand and evaluate the conclusions of this paper are provided. Single-cell RNA sequencing data have been deposited in the Gene Expression Omnibus database under the accession number GSE229976. Bulk RNA sequencing data have been deposited under the accession number GSE247742Source data are provided with this paper.

References

  1. Abraham, C. & Cho, J. H. IL-23 and autoimmunity: new insights into the pathogenesis of inflammatory bowel disease. Annu. Rev. Med. 60, 97–110 (2009).

    CAS  PubMed  Google Scholar 

  2. Maloy, K. J. & Powrie, F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature 474, 298–306 (2011).

    ADS  CAS  PubMed  Google Scholar 

  3. Maynard, C. L., Elson, C. O., Hatton, R. D. & Weaver, C. T. Reciprocal interactions of the intestinal microbiota and immune system. Nature 489, 231–241 (2012).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Fragoulis, G. E., Siebert, S. & McInnes, I. B. Therapeutic targeting of IL-17 and IL-23 cytokines in immune-mediated diseases. Annu. Rev. Med. 67, 337–353 (2016).

    CAS  PubMed  Google Scholar 

  5. Bernink, J. H. et al. Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nat. Immunol. 14, 221–229 (2013).

    CAS  PubMed  Google Scholar 

  6. Takayama, T. et al. Imbalance of NKp44+NKp46 and NKp44NKp46+ natural killer cells in the intestinal mucosa of patients with Crohn’s disease. Gastroenterology 139, 882–892 (2010).

    CAS  PubMed  Google Scholar 

  7. Zhou, L. et al. Innate lymphoid cells support regulatory T cells in the intestine through interleukin-2. Nature 568, 405–409 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Oppmann, B. et al. Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity 13, 715–725 (2000).

    CAS  PubMed  Google Scholar 

  9. Neurath, M. F. IL-23: a master regulator in Crohn disease. Nat. Med. 13, 26–28 (2007).

    CAS  PubMed  Google Scholar 

  10. Krueger, G. G. et al. A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis. N. Engl. J. Med. 356, 580–592 (2007).

    CAS  PubMed  Google Scholar 

  11. Lubberts, E. The IL-23-IL-17 axis in inflammatory arthritis. Nat. Rev. Rheumatol. 11, 415–429 (2015).

    CAS  PubMed  Google Scholar 

  12. Cua, D. J. et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature 421, 744–748 (2003).

    ADS  CAS  PubMed  Google Scholar 

  13. Duerr, R. H. et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 314, 1461–1463 (2006).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. The Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661–678 (2007).

    PubMed Central  Google Scholar 

  15. D’Haens, G. et al. Risankizumab as induction therapy for Crohn’s disease: results from the phase 3 ADVANCE and MOTIVATE induction trials. Lancet 399, 2015–2030 (2022).

    PubMed  Google Scholar 

  16. Feagan, B. G. et al. Ustekinumab as induction and maintenance therapy for Crohn’s disease. N. Engl. J. Med. 375, 1946–1960 (2016).

    CAS  PubMed  Google Scholar 

  17. Sands, B. E. et al. Ustekinumab as induction and maintenance therapy for ulcerative colitis. N. Engl. J. Med. 381, 1201–1214 (2019).

    CAS  PubMed  Google Scholar 

  18. Shih, V. F. et al. Homeostatic IL-23 receptor signaling limits Th17 response through IL-22-mediated containment of commensal microbiota. Proc. Natl Acad. Sci. USA 111, 13942–13947 (2014).

    ADS  PubMed  PubMed Central  Google Scholar 

  19. Mangan, P. R. et al. Transforming growth factor-β induces development of the TH17 lineage. Nature 441, 231–234 (2006).

    ADS  CAS  PubMed  Google Scholar 

  20. Zheng, Y. et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat. Med. 14, 282–289 (2008).

    CAS  PubMed  Google Scholar 

  21. Sonnenberg, G. F., Monticelli, L. A., Elloso, M. M., Fouser, L. A. & Artis, D. CD4+ lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity 34, 122–134 (2011).

    CAS  PubMed  Google Scholar 

  22. Cox, J. H. et al. Opposing consequences of IL-23 signaling mediated by innate and adaptive cells in chemically induced colitis in mice. Mucosal Immunol. 5, 99–109 (2012).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Martínez-Barricarte, R. et al. Human IFN-γ immunity to mycobacteria is governed by both IL-12 and IL-23. Sci. Immunol. 3, eaau6759 (2018).

    PubMed  Google Scholar 

  25. Mao, K. et al. Innate and adaptive lymphocytes sequentially shape the gut microbiota and lipid metabolism. Nature 554, 255–259 (2018).

    ADS  CAS  PubMed  Google Scholar 

  26. Sawa, S. et al. RORγt+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nat. Immunol. 12, 320–326 (2011).

    CAS  PubMed  Google Scholar 

  27. Sonnenberg, G. F. & Artis, D. Innate lymphoid cells in the initiation, regulation and resolution of inflammation. Nat. Med. 21, 698–708 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Spits, H. et al. Innate lymphoid cells—a proposal for uniform nomenclature. Nat. Rev. Immunol. 13, 145–149 (2013).

    CAS  PubMed  Google Scholar 

  29. Kerdiles, Y. M. et al. Foxo transcription factors control regulatory T cell development and function. Immunity 33, 890–904 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Hossain, D. M. et al. FoxP3 acts as a cotranscription factor with STAT3 in tumor-induced regulatory T cells. Immunity 39, 1057–1069 (2013).

    CAS  PubMed  Google Scholar 

  31. Jacobse, J. et al. Interleukin-23 receptor signaling impairs the stability and function of colonic regulatory T cells. Cell Rep. 42, 112128 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Kannan, A. K. et al. IL-23 induces regulatory T cell plasticity with implications for inflammatory skin diseases. Sci. Rep. 9, 17675 (2019).

    ADS  PubMed  PubMed Central  Google Scholar 

  33. Uhlig, H. H. et al. Differential activity of IL-12 and IL-23 in mucosal and systemic innate immune pathology. Immunity 25, 309–318 (2006).

    CAS  PubMed  Google Scholar 

  34. Ahern, P. P. et al. Interleukin-23 drives intestinal inflammation through direct activity on T cells. Immunity 33, 279–288 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Wing, K. et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science 322, 271–275 (2008).

    ADS  CAS  PubMed  Google Scholar 

  36. Walker, L. S. & Sansom, D. M. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nat. Rev. Immunol. 11, 852–863 (2011).

    CAS  PubMed  Google Scholar 

  37. Chang, T. T., Jabs, C., Sobel, R. A., Kuchroo, V. K. & Sharpe, A. H. Studies in B7-deficient mice reveal a critical role for B7 costimulation in both induction and effector phases of experimental autoimmune encephalomyelitis. J. Exp. Med. 190, 733–740 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Lenschow, D. J. et al. CD28/B7 regulation of Th1 and Th2 subsets in the development of autoimmune diabetes. Immunity 5, 285–293 (1996).

    CAS  PubMed  Google Scholar 

  39. Tekguc, M., Wing, J. B., Osaki, M., Long, J. & Sakaguchi, S. Treg-expressed CTLA-4 depletes CD80/CD86 by trogocytosis, releasing free PD-L1 on antigen-presenting cells. Proc. Natl Acad. Sci. USA 118, e2023739118 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Sugiura, D. et al. Restriction of PD-1 function by cis-PD-L1/CD80 interactions is required for optimal T cell responses. Science 364, 558–566 (2019).

    ADS  CAS  PubMed  Google Scholar 

  41. Francisco, L. M. et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J. Exp. Med. 206, 3015–3029 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Keir, M. E. et al. Tissue expression of PD-L1 mediates peripheral T cell tolerance. J. Exp. Med. 203, 883–895 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Lyu, M. et al. ILC3s select microbiota-specific regulatory T cells to establish tolerance in the gut. Nature 610, 744–751 (2022).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Paustian, A. M. S. et al. Continuous IL-23 stimulation drives ILC3 depletion in the upper GI tract and, in combination with TNFα, induces robust activation and a phenotypic switch of ILC3. PLoS ONE 12, e0182841 (2017).

    PubMed  PubMed Central  Google Scholar 

  45. Jacquelot, N. et al. Immune checkpoints and innate lymphoid cells—new avenues for cancer immunotherapy. Cancers (Basel) 13, 5967 (2021).

    CAS  PubMed  Google Scholar 

  46. Hepworth, M. R. et al. Immune tolerance. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4+ T cells. Science 348, 1031–1035 (2015).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  47. Goc, J. et al. Dysregulation of ILC3s unleashes progression and immunotherapy resistance in colon cancer. Cell 184, 5015–5030.e5016 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Kløverpris, H. N. et al. Innate lymphoid cells are depleted irreversibly during acute HIV-1 infection in the absence of viral suppression. Immunity 44, 391–405 (2016).

    PubMed  PubMed Central  Google Scholar 

  49. Page, D. B., Postow, M. A., Callahan, M. K., Allison, J. P. & Wolchok, J. D. Immune modulation in cancer with antibodies. Annu. Rev. Med. 65, 185–202 (2014).

    CAS  PubMed  Google Scholar 

  50. Lo, B. C. et al. Microbiota-dependent activation of CD4+ T cells induces CTLA-4 blockade-associated colitis via Fcγ receptors. Science 383, 62–70 (2024).

    ADS  CAS  PubMed  Google Scholar 

  51. Mombaerts, P. et al. RAG-1-deficient mice have no mature B and T lymphocytes. Cell 68, 869–877 (1992).

    CAS  PubMed  Google Scholar 

  52. Kühn, R., Löhler, J., Rennick, D., Rajewsky, K. & Müller, W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 75, 263–274 (1993).

    PubMed  Google Scholar 

  53. Awasthi, A. et al. Cutting edge: IL-23 receptor gfp reporter mice reveal distinct populations of IL-17-producing cells. J. Immunol. 182, 5904–5908 (2009).

    CAS  PubMed  Google Scholar 

  54. Lochner, M. et al. In vivo equilibrium of proinflammatory IL-17+ and regulatory IL-10+ Foxp3+ RORγt+ T cells. J. Exp. Med. 205, 1381–1393 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Narni-Mancinelli, E. et al. Fate mapping analysis of lymphoid cells expressing the NKp46 cell surface receptor. Proc. Natl Acad. Sci. USA 108, 18324–18329 (2011).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  56. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Sonnenberg Laboratory for discussions and critical reading of the manuscript. Research in the Sonnenberg Laboratory is supported by the US National Institutes of Health (grant nos. R01AI143842, R01AI123368, R01AI145989, U01AI095608, R01AI162936, R01CA274534 and R37AI174468), an Investigators in the Pathogenesis of Infectious Disease Award from the Burroughs Wellcome Fund, the Meyer Cancer Center Collaborative Research Initiative, the Dalton Family Foundation and Linda and Glenn Greenberg. A.A. and M.L. are supported by fellowships from the Crohn’s and Colitis Foundation (grant nos. 902451 and 935259). A.M.J. and J.U. are supported by US National Institutes of Health grant no. T32DK116970. J.Z. is supported by grant no. F32DK136248. V.H. is supported by a DFG Walter Benjamin Fellowship (grant no. HO 7399/1-1). G.F.S. is a CRI Lloyd J. Old STAR. We thank the Epigenomics Cores of Weill Cornell Medicine, N. Zahan for administrative support and D. Garone and A. Brcic-Susak for technical support. The Jill Roberts Institute (JRI) Live Cell Bank is supported by the JRI, the Jill Roberts Center for IBD, Cure for IBD, the Rosanne H. Silbermann Foundation, the Sanders Family and Weill Cornell Medicine Division of Pediatric Gastroenterology and Nutrition.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

A.A. and G.F.S. conceived the project. A.A. performed most experiments and analysed the data. A.M.J., J.Z., V.H., J.U., M.L. and J.G. helped with experiments and data analyses. JRI Live Cell Bank and R.E.S. contributed to clinical sample acquisition, annotation, processing and evaluation. J.B.W., E.V. and S.S. provided essential mouse models, scientific advice and expertise. A.A. and G.F.S. wrote the manuscript, with input from all of the authors.

Corresponding author

Correspondence to Gregory F. Sonnenberg.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewers 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 Single cell analysis of all IL-23R+ cells in small intestine of healthy mice.

a, Gating strategy and experimental design to sort IL-23R+ immune cells from small intestine lamina propria of IL-23R-eGFP mice for scRNA-seq. Violin plot confirming Il23r expression among all the identified IL-23R+ cell clusters. b, Feature plots showing expression of indicated genes in different clusters. c, Violin plot showing Mki67 expression among all identified IL-23R+ cell clusters.

Extended Data Fig. 2 Characterization of IL-23R+ immune cells in the intestine.

a, Flow cytometry plots with donut plot of final frequencies of different IL-23R+ immune cells in small intestine lamina propria of IL-23R-eGFP mice (n = 4 mice). Lineage 1, CD11b, CD11c, B220, CD3ε, CD5, CD8α, NK1.1 and TCRγδ. b, Flow cytometry plots showing eGFP expression (IL-23R) in different immune cells in small intestine lamina propria of IL-23R-eGFP mice (n = 4 mice). c, Donut plots of final frequencies of different IL-23R+ immune cells in large intestine lamina propria of naïve and C. rodentium infected IL-23R-eGFP mice at day 14 post infection (n = 4 mice). Data in a and c are representative of two or three independent experiments with similar results.

Source Data

Extended Data Fig. 3 Acute IL-23 driven responses in IL-23R+ intestinal cells.

a, Volcano plots of differentially expressed genes in scRNA-seq dataset from IL-23R+ immune cells from the small intestine of IL-23R-eGFP mice before and after IL-23 stimulation in annotated cell types. b, UMAP plots of scRNA-seq data showing indicated gene expression in different clusters before and after IL-23 stimulation (1-NKp46+ ILC3s, 2-CCR6+ ILC3s, 3-DN ILC3s, 4-γδ T cells, 5-TH17 cells, 6-Proliferative cells). The statistics was obtained by the Wilcoxon test as implemented by Seurat; red dots are significantly different.

Extended Data Fig. 4 Flow cytometry gating for ILCs and CTLA-4.

a, Gating strategy for flow cytometry analysis of different immune cells in intestinal lamina propria of mice. Lineage 1, CD11b, CD11c, F4/80 and B220; lineage 2, CD3ε, CD5, CD8α and TCRγδ; Group 1 innate lymphoid cells (ILC1s) were identified as live CD45+ Lineage CD127+ CD90.2+ T-bet+ RORγ NK1.1+ Eomes; group 2 innate lymphoid cells (ILC2s) were identified as live CD45+ Lineage CD127+ CD90.2+ GATA3+; group 3 innate lymphoid cells (ILC3s) were identified as live CD45+ Lineage CD127+ CD90.2+ RORγ+; and subsets of ILC3s were further identified as CCR6+ T-bet ILC3s or CCR6 T-bet+ ILC3s. For T cell analysis Tregs were identified as CD45+CD4+FoxP3+ T cells and TH 17 as CD45+CD4+ RORγ+FoxP3 T cells. b, Representative flow cytometry plots of CTLA-4 staining in different immune cells from small intestine lamina propria of C57BL/6 mice (n = 4 mice). c, Flow cytometry plots of CTLA-4+ cells in small intestine lamina propria of Rag1−/− mice (n = 4 mice). Data in b and c are representative of two independent experiments with similar results.

Extended Data Fig. 5 Microbiota dependent regulation of CTLA-4.

a, Flow cytometry plots with graph of CTLA-4+ ILC1s and CTLA-4+ NK cells frequencies in large intestine lamina propria of conventional SPF, germ-free (GF) and SPF Rag1−/− mice treated with antibiotics (ABX) (n = 8 mice). b, Graph of CTLA-4+ ILC1s and CTLA-4+ NK cells frequencies in small intestine lamina propria of conventional SPF, germ-free and SPF mice treated with antibiotics on Rag1−/− background (n = 8 mice). c, Graph of CTLA-4+ ILC1s, CTLA-4+ NK cells and CTLA-4+ γδ T cells frequencies in small intestine lamina propria of SPF mice ex vivo stimulated with indicated cytokines (n = 4 mice). Data in a-b were pooled from two independent experiments and shown as Mean ± s.d. Data in c are representative of two or three independent experiments with similar results and shown as Mean ± s.d. The statistics in a-c were calculated by one-way ANOVA with Dunnett’s multiple comparisons.

Source Data

Extended Data Fig. 6 Steady state analysis of Ncr1cre Ctla4f/f mice.

a, Flow cytometry plots of NKp46+ cells in large intestine lamina propria of naïve and C. rodentium infected C57BL/6 mice (n = 4 mice). Lineage 1, CD11b, CD11c and B220; lineage 2, CD3ε, CD5, CD8α, and TCRγδ. b, UMAP plot of scRNA-seq data of IL-23R+ immune cells from the small intestine of IL-23R-eGFP mice showing Ctla4 expression in different clusters. The large intestine of Ncr1cre Ctla4f/f and Ctla4f/f were analyzed at steady state and graph displaying the frequency of (c) ILC3s (percentage of Lin- CD90+ CD127+) and (d) IL-22+ ILC3s (percentage of total ILC3s). e, Graph of TH17 cells frequency (percentage of CD4+ T cells). Flow plots with graph displaying the frequency of (f) Treg cells (percentage of CD4+ T cells) and (g) neutrophils gated as CD11b+Ly6G+ cells (percentage of CD45+ cells). Data in a,c,d,e,f and g are representative of two or three independent experiments with similar results (n = 5 mice) and shown as Mean ± s.d. The statistics in c-g were determined by Mann–Whitney U-test (unpaired, two-tailed).

Source Data

Extended Data Fig. 7 NK cells and ILC1s do not impact intestinal inflammation following enteric infection with C. rodentium.

C57BL/6 mice were orally infected with C. rodentium and treated with IgG or anti-IL-23 monoclonal antibody. a, Flow cytometry plots with graph displaying the frequency of CTLA-4 expression on ILC1s, NK cells and γδ T cells (n = 5 mice). C57BL/6 mice were orally infected with C. rodentium and treated with IgG or anti-NK1.1 monoclonal antibody. Mice were analyzed at day 14 post infection and graph displaying the (b) depletion efficiency of natural killer cells and ILC1s, and the frequency of (c) ILC3s (percentage of Lin CD90+ CD127+); (d) Treg cells (percentage of CD4+ T cells); (e) TH17 cells (percentage of CD4+ T cells); (f) Neutrophils gated as CD11b+Ly6G+ cells (percentage of CD45+ cells) in large intestinal lamina propria immune cells. Data in b-f are representative of two or three independent experiments with similar results (n = 4 mice) and shown as Mean ± s.d. The statistics in a-f were determined by Mann–Whitney U-test (unpaired, two-tailed).

Source Data

Extended Data Fig. 8 Innate immune responses are intact in mice lacking ILC3-specific CTLA-4.

Ncr1wt/wt and Ncr1cre/wt mice were orally infected with C. rodentium and at day 14 post infection large intestinal lamina propria immune cells were analyzed. a, Flow cytometry plots with graph of Tregs frequency (percentage of CD4+ T cells) and graph displaying the frequency of (b) TH17 cells (percentage of CD4+ T cells) and (c) neutrophils gated as CD11b+Ly6G+ cells (percentage of CD45+ cells) with (d) colon length. e, Flow cytometry plots and graph of CTLA-4+ ILC3s frequency (percentage of total ILC3s) in Rag1−/− Ctla4f/f and Rag1−/− RORγtcre Ctla4f/f mice. Rag1−/− Ctla4f/f and Rag1−/− RORγtcre Ctla4f/f mice were treated with 100 µg of anti-CD40 antibody, and graph displaying the frequency of (f) ILC3s (percentage of Lin CD90+ CD127+); (g) IL-22+ ILC3s (percentage of total ILC3s) and (h) neutrophils gated as CD11b+Ly6G+ cells (percentage of CD45+ cells) in the large intestines with (i) colon length at day 7 post treatment. Data in a-i are representative of two independent experiments with similar results (n = 4 mice) and shown as Mean ± s.d. The statistics in a-i were determined by Mann–Whitney U-test (unpaired, two-tailed).

Source Data

Extended Data Fig. 9 CTLA-4+ ILC3s reduce co-stimulatory molecules and enhance PD-L1 on myeloid cells in response to IL-23.

Sorted ILC3s and myeloid cells from the large intestine of C57BL/6 mice were co-cultured with recombinant IL-23 in presence or absence of indicated blocking antibodies. Flow cytometry plots with graph of MFI of (a) CD80 and (b) CD86 expression on myeloid cells gated as CD11c+ MHCIIhi. c, Flow cytometry plots with graph of free PD-L1 frequency on myeloid cells gated as CD11c+ MHCIIhi. d, Gating strategy to analyze T cells, myeloid cells and ILC3s in human samples. Data in a-c are representative of two independent experiments with similar results (n = 4 mice) and shown as Mean ± s.d. The statistics were calculated by one-way ANOVA with Dunnett’s multiple comparisons.

Source Data

Extended Data Fig. 10 ILC3s restrain IL-23 mediated inflammation through CTLA-4.

Here we define a pathway of immune regulation in the large intestine. This pathway is activated by gut microbes and IL-23 in a FOXO1- and STAT3-dependent manner. ILC3-intrinsic CTLA-4 shapes the levels of co-inhibitory and co-stimulatory molecules on intestinal myeloid cells to support Tregs and restrict T effector (Teff) cells. Consequently, ILC3-intrinsic CTLA-4 function as a checkpoint to restrain the pathologic functions of IL-23, suggesting that disruption of these lymphocytes, which occurs in IBD, contributes to chronic inflammation.

Supplementary information

Reporting Summary

Supplementary Table 1

Clinical metadata on human samples for bulk RNA sequencing.

Supplementary Table 2

Clinical metadata on paediatric patients with IBD and matched healthy controls.

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

Ahmed, A., Joseph, A.M., Zhou, J. et al. CTLA-4-expressing ILC3s restrain interleukin-23-mediated inflammation. Nature 630, 976–983 (2024). https://doi.org/10.1038/s41586-024-07537-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07537-3

Comments

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

Search

Quick links

Nature Briefing

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

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