Close banner

2022-09-10 11:02:03 By : Mr. OLIVER CHEN

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.

Microbial colonization of the mammalian intestine elicits inflammatory or tolerogenic T cell responses, but the mechanisms controlling these distinct outcomes remain poorly understood, and accumulating evidence indicates that aberrant immunity to intestinal microbiota is causally associated with infectious, inflammatory and malignant diseases1,2,3,4,5,6,7,8. Here we define a critical pathway controlling the fate of inflammatory versus tolerogenic T cells that respond to the microbiota and express the transcription factor RORγt. We profiled all RORγt+ immune cells at single-cell resolution from the intestine-draining lymph nodes of mice and reveal a dominant presence of T regulatory (Treg) cells and lymphoid tissue inducer-like group 3 innate lymphoid cells (ILC3s), which co-localize at interfollicular regions. These ILC3s are distinct from extrathymic AIRE-expressing cells, abundantly express major histocompatibility complex class II, and are necessary and sufficient to promote microbiota-specific RORγt+ Treg cells and prevent their expansion as inflammatory T helper 17 cells. This occurs through ILC3-mediated antigen presentation, αV integrin and competition for interleukin-2. Finally, single-cell analyses suggest that interactions between ILC3s and RORγt+ Treg cells are impaired in inflammatory bowel disease. Our results define a paradigm whereby ILC3s select for antigen-specific RORγt+ Treg cells, and against T helper 17 cells, to establish immune tolerance to the microbiota and intestinal health.

Your institute does not have access to this article

Get full journal access for 1 year

All prices are NET prices. VAT will be added later in the checkout. Tax calculation will be finalised during checkout.

Get time limited or full article access on ReadCube.

All prices are NET prices.

All data necessary to understand and evaluate the conclusions of this paper are provided in the manuscript and supplementary materials. scRNA-seq data have been deposited in the Gene Expression Omnibus database under the accession numbers GSE184175 and GSE184291. A reanalysis was performed on publicly available scRNA-seq data with the accession number GSE134809. Source data are provided with this paper.

Honda, K. & Littman, D. R. The microbiota in adaptive immune homeostasis and disease. Nature 535, 75–84 (2016).

ADS  CAS  PubMed  Google Scholar 

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

ADS  CAS  PubMed  Google Scholar 

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 

Zhou, L. & Sonnenberg, G. F. Essential immunologic orchestrators of intestinal homeostasis. Sci. Immunol. 3, eaao1605 (2018).

PubMed  PubMed Central  Google Scholar 

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

CAS  PubMed  PubMed Central  Google Scholar 

Hooper, L. V. & Macpherson, A. J. Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat. Rev. Immunol. 10, 159–169 (2010).

Harrison, O. J. & Powrie, F. M. Regulatory T cells and immune tolerance in the intestine. Cold Spring Harb. Perspect. Biol. 5, a018341 (2013).

PubMed  PubMed Central  Google Scholar 

Janney, A., Powrie, F. & Mann, E. H. Host–microbiota maladaptation in colorectal cancer. Nature 585, 509–517 (2020).

ADS  CAS  PubMed  Google Scholar 

Harrington, L. E. et al. Interleukin 17–producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat. Immunol. 6, 1123–1132 (2005).

Park, H. et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat. Immunol. 6, 1133–1141 (2005).

CAS  PubMed  PubMed Central  Google Scholar 

Ohnmacht, C. et al. The microbiota regulates type 2 immunity through RORγt+ T cells. Science 349, 989–993 (2015).

ADS  CAS  PubMed  Google Scholar 

Sefik, E. et al. Individual intestinal symbionts induce a distinct population of RORγ+ regulatory T cells. Science 349, 993–997 (2015).

ADS  CAS  PubMed  PubMed Central  Google Scholar 

Buonocore, S. et al. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature 464, 1371–1375 (2010).

ADS  CAS  PubMed  PubMed Central  Google Scholar 

Cella, M. et al. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature 457, 722–725 (2009).

ADS  CAS  PubMed  Google Scholar 

Sawa, S. et al. Lineage relationship analysis of RORγt+ innate lymphoid cells. Science 330, 665–669 (2010).

ADS  CAS  PubMed  Google Scholar 

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

Eberl, G. RORγt, a multitask nuclear receptor at mucosal surfaces. Mucosal Immunol. 10, 27–34 (2017).

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

Yamano, T. et al. Aire-expressing ILC3-like cells in the lymph node display potent APC features. J. Exp. Med. 216, 1027–1037 (2019).

CAS  PubMed  PubMed Central  Google Scholar 

Cabeza-Cabrerizo, M., Cardoso, A., Minutti, C. M., Pereira da Costa, M. & Reis e Sousa, C. Dendritic cells revisited. Annu. Rev. Immunol. 39, 131–166 (2021).

Zhou, W. et al. ZBTB46 defines and regulates ILC3s that protect the intestine. Nature 609, 159–165 (2022).

Gardner, J. M. et al. Extrathymic Aire-expressing cells are a distinct bone marrow-derived population that induce functional inactivation of CD4+ T cells. Immunity 39, 560–572 (2013).

CAS  PubMed  PubMed Central  Google Scholar 

Wells, K. L. et al. Combined transient ablation and single-cell RNA-sequencing reveals the development of medullary thymic epithelial cells. eLife 9, e60188 (2020).

CAS  PubMed  PubMed Central  Google Scholar 

Anderson, M. S. et al. Projection of an immunological self shadow within the thymus by the aire protein. Science 298, 1395–1401 (2002).

ADS  CAS  PubMed  Google Scholar 

Mackley, E. C. et al. CCR7-dependent trafficking of RORγ+ ILCs creates a unique microenvironment within mucosal draining lymph nodes. Nat. Commun. 6, 5862 (2015).

ADS  CAS  PubMed  Google Scholar 

Hepworth, M. R. et al. 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 

Coombes, J. L. et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J. Exp. Med. 204, 1757–1764 (2007).

CAS  PubMed  PubMed Central  Google Scholar 

Farache, J. et al. Luminal bacteria recruit CD103+ dendritic cells into the intestinal epithelium to sample bacterial antigens for presentation. Immunity 38, 581–595 (2013).

CAS  PubMed  PubMed Central  Google Scholar 

Sun, C. M. et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J. Exp. Med. 204, 1775–1785 (2007).

CAS  PubMed  PubMed Central  Google Scholar 

Tanoue, T., Atarashi, K. & Honda, K. Development and maintenance of intestinal regulatory T cells. Nat. Rev. Immunol. 16, 295–309 (2016).

Russler-Germain, E. V. et al. Gut Helicobacter presentation by multiple dendritic cell subsets enables context-specific regulatory T cell generation. eLife 10, e54792 (2021).

Hepworth, M. R. et al. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature 498, 113–117 (2013).

ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

ADS  CAS  PubMed  PubMed Central  Google Scholar 

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 

Belkaid, Y. & Oldenhove, G. Tuning microenvironments: induction of regulatory T cells by dendritic cells. Immunity 29, 362–371 (2008).

CAS  PubMed  PubMed Central  Google Scholar 

Worthington, J. J., Czajkowska, B. I., Melton, A. C. & Travis, M. A. Intestinal dendritic cells specialize to activate transforming growth factor-β and induce Foxp3+ regulatory T cells via integrin αvβ8. Gastroenterology 141, 1802–1812 (2011).

Worthington, J. J. et al. Integrin αvβ8-mediated TGF-β activation by effector regulatory T cells is essential for suppression of T-cell-mediated inflammation. Immunity 42, 903–915 (2015).

CAS  PubMed  PubMed Central  Google Scholar 

Ouyang, W., Beckett, O., Ma, Q. & Li, M. O. Transforming growth factor-beta signaling curbs thymic negative selection promoting regulatory T cell development. Immunity 32, 642–653 (2010).

CAS  PubMed  PubMed Central  Google Scholar 

Sarrazy, V. et al. Integrins αvβ5 and αvβ3 promote latent TGF-β1 activation by human cardiac fibroblast contraction. Cardiovasc. Res. 102, 407–417 (2014).

CAS  PubMed  PubMed Central  Google Scholar 

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

Teng, F. et al. A circadian clock is essential for homeostasis of group 3 innate lymphoid cells in the gut. Sci. Immunol. 4, eaax1215 (2019).

CAS  PubMed  PubMed Central  Google Scholar 

Vivier, E. et al. Innate lymphoid cells: 10 years on. Cell 174, 1054–1066 (2018).

Martin, J. C. et al. Single-cell analysis of Crohn's disease lesions identifies a pathogenic cellular module associated with resistance to anti-TNF therapy. Cell 178, 1493–1508 (2019).

CAS  PubMed  PubMed Central  Google Scholar 

Hovhannisyan, Z., Treatman, J., Littman, D. R. & Mayer, L. Characterization of interleukin-17-producing regulatory T cells in inflamed intestinal mucosa from patients with inflammatory bowel diseases. Gastroenterology 140, 957–965 (2011).

Jaeger, N. et al. Single-cell analyses of Crohn's disease tissues reveal intestinal intraepithelial T cells heterogeneity and altered subset distributions. Nat. Commun. 12, 1921 (2021).

ADS  PubMed  PubMed Central  Google Scholar 

Boland, B. S. et al. Heterogeneity and clonal relationships of adaptive immune cells in ulcerative colitis revealed by single-cell analyses. Sci. Immunol. 5, eabb4432 (2020).

CAS  PubMed  PubMed Central  Google Scholar 

Smillie, C. S. et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell 178, 714–730 (2019).

CAS  PubMed  PubMed Central  Google Scholar 

Friedrich, M., Pohin, M. & Powrie, F. Cytokine networks in the pathophysiology of inflammatory bowel disease. Immunity 50, 992–1006 (2019).

Grigg, J. B. et al. Antigen-presenting innate lymphoid cells orchestrate neuroinflammation. Nature 600, 707–712 (2021).

ADS  CAS  PubMed  PubMed Central  Google Scholar 

Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4 (2001).

CAS  PubMed  PubMed Central  Google Scholar 

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

Nussbaum, J. C. et al. Type 2 innate lymphoid cells control eosinophil homeostasis. Nature 502, 245–248 (2013).

ADS  CAS  PubMed  PubMed Central  Google Scholar 

Rubtsov, Y. P. et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546–558 (2008).

Lee, P. P. et al. A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity 15, 763–774 (2001).

Caton, M. L., Smith-Raska, M. R. & Reizis, B. Notch–RBP-J signaling controls the homeostasis of CD8− dendritic cells in the spleen. J. Exp. Med. 204, 1653–1664 (2007).

CAS  PubMed  PubMed Central  Google Scholar 

Schraml, B. U. et al. Genetic tracing via DNGR-1 expression history defines dendritic cells as a hematopoietic lineage. Cell 154, 843–858 (2013).

Ahlfors, H. et al. IL-22 fate reporter reveals origin and control of IL-22 production in homeostasis and infection. J. Immunol. 193, 4602–4613 (2014).

Choi, G. B. et al. The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science 351, 933–939 (2016).

ADS  CAS  PubMed  PubMed Central  Google Scholar 

Lacy-Hulbert, A. et al. Ulcerative colitis and autoimmunity induced by loss of myeloid αv integrins. Proc. Natl Acad. Sci. USA 104, 15823–15828 (2007).

ADS  CAS  PubMed  PubMed Central  Google Scholar 

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 

Fan, Y. et al. Thymus-specific deletion of insulin induces autoimmune diabetes. EMBO J. 28, 2812–2824 (2009).

CAS  PubMed  PubMed Central  Google Scholar 

Archambault, A. S. et al. Cutting edge: conditional MHC class II expression reveals a limited role for B cell antigen presentation in primary and secondary CD4 T cell responses. J. Immunol. 191, 545–550 (2013).

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 

Schlenner, S. M. et al. Fate mapping reveals separate origins of T cells and myeloid lineages in the thymus. Immunity 32, 426–436 (2010).

Popmihajlov, Z., Xu, D., Morgan, H., Milligan, Z. & Smith, K. A. Conditional IL-2 gene deletion: consequences for T cell proliferation. Front. Immunol. 3, 102 (2012).

CAS  PubMed  PubMed Central  Google Scholar 

Withers, D. R. et al. Cutting edge: lymphoid tissue inducer cells maintain memory CD4 T cells within secondary lymphoid tissue. J. Immunol. 189, 2094–2098 (2012).

Kim, S. et al. CD117+ CD3− CD56− OX40Lhigh cells express IL-22 and display an LTi phenotype in human secondary lymphoid tissues. Eur. J. Immunol. 41, 1563–1572 (2011).

Dutton, E. E. & Withers, D. R. Identification of murine and human innate lymphoid cells in frozen tissue sections using immunofluorescence. Methods Mol. Biol. 2121, 51–58 (2020).

Yang, B. H. et al. Foxp3+ T cells expressing RORγt represent a stable regulatory T-cell effector lineage with enhanced suppressive capacity during intestinal inflammation. Mucosal Immunol. 9, 444–457 (2016).

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 

Hadley, W. ggplot2: Elegant Graphics for Data Analysis (Springer, 2016).

von Mering, C. et al. STRING: known and predicted protein–protein associations, integrated and transferred across organisms. Nucleic Acids Res. 33, D433–D437 (2005).

We thank members of the Sonnenberg Laboratory for discussions and critical reading of the manuscript. Research in the Sonnenberg Laboratory is supported by the National Institutes of Health (R01AI143842, R01AI123368, R01AI145989, U01AI095608, R21CA249274, R01AI162936 and R01CA274534), 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. W. Zhou, J.G., L.Z. and W. Zhang are supported by fellowships from the Crohn’s and Colitis Foundation (831404, 519428, 608975 and 901000, respectively). D.R.W. and F.G. are supported by a Senior Research Fellowship from the Wellcome Trust to D.R.W. (110199/Z/15/Z). J.G.F. is supported by P30-ES002109 and R35CA210088. G.F.S. is a CRI Lloyd J. Old STAR. We thank the Epigenomics Cores of Weill Cornell Medicine and G. Putzel for bioinformatics assistance, J. Conrad for administrative assistance, and S. Mozumder for technical assistance. The JRI IBD 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, Hepatology, and Nutrition.

Present address: EA Pharma, Kanagawa, Japan

These authors contributed equally: Mengze Lyu, Hiroaki Suzuki

Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA

Mengze Lyu, Hiroaki Suzuki, Lan Kang, Wenqing Zhou, Jeremy Goc, Lei Zhou, Jordan Zhou, Wen Zhang, Gregory F. Sonnenberg, Wenqing Zhou & Gregory F. Sonnenberg

Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA

Mengze Lyu, Hiroaki Suzuki, Lan Kang, Wenqing Zhou, Jeremy Goc, Lei Zhou, Jordan Zhou, Wen Zhang, Gregory F. Sonnenberg, Wenqing Zhou & Gregory F. Sonnenberg

Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA

Mengze Lyu, Hiroaki Suzuki, Lan Kang, Wenqing Zhou, Jeremy Goc, Lei Zhou, Jordan Zhou, Wen Zhang, David Artis, Randy Longman, Gregory F. Sonnenberg, Ellen Scherl, Dana Lukin, Robert Battat, Thomas Ciecierega, Aliza Solomon, Elaine Barfield, Kimberley Chien, Johanna Ferriera, Jasmin Williams, Shaira Khan, Peik Sean Chong, Samah Mozumder, Lance Chou, Wenqing Zhou, Anees Ahmed, Connie Zhong, Ann Joseph, Sanchita Kashyap, Joseph Gladstone, Samantha Jensen & Gregory F. Sonnenberg

Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK

Fabrina Gaspal & David R. Withers

Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA

Zeli Shen & James G. Fox

Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Weill Cornell Medicine, Cornell University, New York, NY, USA

Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA

Robbyn E. Sockolow & Terri M. Laufer

Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, USA

Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, USA

Microenvironment and Immunity Unit, Institut Pasteur, Paris, France

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

You can also search for this author in PubMed  Google Scholar

M.L., H.S. and G.F.S. conceived the project. M.L. performed most experiments and analysed the data. H.S., L.K., F.G., J.G., W. Zhou, L.Z., W. Zhang and J.Z. helped with experiments and data analyses. J.G.F., Z.S., Y.F., T.M.L., G.E. and D.R.W. provided essential tools, scientific advice and expertise. R.E.S. and JRI Live Cell Bank contributed to clinical sample acquisition and processing. M.L. and G.F.S. wrote the manuscript, with input from all authors.

Correspondence to Gregory F. Sonnenberg.

H.S. is currently employed by EA Pharma. The other authors declare no competing interests.

Nature thanks the anonymous reviewers for their contribution to the peer review of this work.

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

a, Gating strategy to sort GFP+TCRβ+ and GFP+TCRβ− cells (1:1, n = 3) for scRNA-seq. b, Doublet test showing cluster 8 as doublets. c, d, Representative flow cytometry plot of the frequency of RORγt+ cells in CD127+ ILC fractions (CD45+CD3ε−CD5−NK1.1−Ly6G−TCRγ/δ−B220−CD11b−CD11c−KLRG1−CD127+), CD19+ B cell fractions (CD45+CD19+) and CD172a+ cDC2 fractions (CD45+ CD3ε−CD5−NK1.1−Ly6G−TCRγ/δ−B220−CD64−CD11c+MHCII+XCR1−CD172a+) from RORγt-eGFP reporter mice (n = 3) (c) and RorccreRosa26lsl−YFP fate mapped mice (n = 3) (d). e–k, Violin plot showing the expression of Rorc (e), Cd3e (f), Foxp3 (g), Xcr1 (h), Clec9a (i), Clec10a (j), Clec12a (k) among all the identified clusters.

a, Violin plot showing the expression of Siglecg and Dpp4 among all the identified clusters of non-T lymphocytes. b, Gating strategy to identify ILC3s and RORγt+ eTACs from mLN of RORγt-eGFP reporter mice (n = 4) for data shown in Fig. 1e, f. c, Quantification of indicated genes expression in LTi-like cells and RORγt+ eTACs shown in Fig. 1e (n = 4). Data in c are representative of three independent experiments. Data are shown as means ± s.e.m., statistics shown in c are obtained by unpaired Student’s t-test (two-tailed).

a, Gating strategy to identify LTi-like ILC3s and RORγt+ eTACs for fate mapping analyses in Fig. 1g, h. b, Representative flow cytometry plots showing expression of CD127, CD11c, CLEC9A and AIRE among “fate-mapped” LTi-like ILC3s and RORγt+ eTACs in mLN shown in Fig. 1g, h. c, Gating strategy to identify LTi-like ILC3s, RORγt+ eTACs and RORγt+ Tregs of mLN in Fig. 2c–l, Fig. 4e, the same gating strategy applied to the LI-LP. d, Gating strategy to identify H. Hepaticus (Hh)-specific CD4+ T cells in mouse LI-LP.

a, Representative flow cytometry plots of frequency (left) and percentage (right) of RORγt+FoxP3+ Tregs and RORγt+FoxP3− TH17 cells among total RORγt+CD4+ T cells in mLN of WT mice (n = 5). b, Tile-scanned (left) and magnified (right) images of mLN stained for expression of IL7Rα (red), CD3 (blue), FOXP3 (cyan), RORγt (green) and DAPI (grey). c–e, Quantification of percentage of RORγt+ Tregs among total Tregs (c), total numbers per mm2 of ILC3s (d) and RORγt+ Tregs (e) in interfollicular zone of mLN of WT-1 (n = 11 areas), WT-2 (n = 8 areas) and WT-3 (n = 12 areas) mice. f, Tile-scanned images and serial sections of mLN stained for expression of IL7Rα (red), CD11c (blue), FOXP3 (cyan), RORγt (green) and DAPI (grey). Left panel is without IL7Rα and RORγt staining, middle panel is without FOXP3 staining, and right panel is a merge with a magnified image. g, Tile-scanned (left) and magnified (right) images of mLN stained for expression of CD3 (red), FOXP3 (cyan), CD11c (green) and DAPI (grey). Scale bars: 50 μm, 20 μm (in magnified images). Data in a, b, f, g are representative of two independent experiments. Data in c–e are representative of two independent experiments containing a total of 5 mice. Data are shown as means ± s.e.m., statistics shown in a are obtained by unpaired Student’s t-test (two-tailed).

a, UMAP plots of scRNA-seq data showing expression of H2-Ab1, H2-Ab1 and Cd74 are enriched in clusters of LTi-like ILC3s and RORγt+ eTACs across all the identified clusters in mouse mLN. b–e, Cell numbers of CD4+ T cells (b), TH17 cells (c), RORγt−Tregs (d) and RORγt+ Tregs (e) in mLN (upper panel) and LI-LP (lower panel) of H2-Ab1fl/fl and MHCIIΔILC3 mice (n = 9, pooled from two independent experiments). f–k, Large intestine of H2-Ab1fl/fl (n = 6) and Cd4creH2-Ab1fl/fl (MHCIIΔT cell) mice (n = 4) (f, g), H2-Ab1fl/fl (n = 4) and Il5creH2-Ab1fl/fl (MHCIIΔILC2) mice (n = 6) (h, i), Rorcfl/fl (n = 10) and Ncr1creRorcfl/fl mice (n = 9) (j, k) were analyzed. Proportion of MHCII-expressing CD4+ T cells (f), ILC2s (h) and ILC3s (j). Frequency of each subset among CD4+ T cells and RORγt+ Tregs among total Tregs (g, i, k). TH17: Foxp3−RORγt+; Treg: Foxp3+; TH1: Foxp3−RORγ−T-bet+; TH2: Foxp3−RORγt−Gata3+. l, Quantification of RORγt+ Tregs among total CD4+ T cells in LI-LP of Airefl/fl and RorccreAirefl/fl mice (n = 5). m, n, Quantification of eTACs among total CD127−CD90− cells (m) and LTi-like ILC3s among CD45+CCR6+ cells (n) in mLN of Rorcfl/fl and AirecreRorcfl/fl mice (n = 5). o, Quantification of MHCII expression among eTACs (o, left) and LTi-like ILC3s (o, right) in mLN of H2-Ab1fl/fl and AirecreH2-Ab1fl/fl mice (n = 4). p, Quantification of MHCII expression among DCs in mLN of H2-Ab1fl/fl and Clec9acreH2-Ab1fl/fl mice (n = 4). Data are representative of two independent experiments unless otherwise indicated. Data shown as mean ± s.e.m. Statistics in f, h, j, l–p, right of g, i, k are obtained by unpaired Student’s t-test. Statistics shown in left of g, i, k are obtained by multiple unpaired t-test. Statistics are calculated by two-tailed test.

a-c, Sort-purified RORγt+CD4+ T cells and LTi-like ILC3s from LI-LP and mLN (n = 6 or 5 or 3 per group as technical replicates) were co-cultured for 72 h and RORγt+ Tregs were analyzed by flow cytometry. Frequency and cell number of RORγt+ Tregs (a, RORγt+Foxp3+ among CD4+ T cells), MFI of Bim (b) and Nur77 (c) in RORγt+ Tregs. d, e, Dead cells were quantified in RORγt+ Tregs (d) and TH17 cells (e) after co-culture with or without LTi-like ILC3s for 72 h. RORγt+CD4+ T cells and LTi-like ILC3s were sort-purified from mLN and LI-LP and pooled for co-culture assay (n = 12, each dot represents samples pooled from 2 mice). Data in a–c are representative of two independent experiments. Data in d, e are pooled from two independent experiments. Data are shown as mean ± s.e.m., statistics shown in d, e were obtained by unpaired Student’s t-test (two-tailed).

a, H. hepaticus (Hh)-specific and/or SFB-specific CD4+ T cells were transferred to H2-Ab1fl/fl and MHCIIΔILC3 mice colonized with H. hepaticus 2 weeks before experiment as shown in Fig. 3a–h, k, l. b, c, Frequency of CD44+ ratio among SFB-specific (b) or Hh-specific (c) CD4+ T cells were analyzed in Peyer’s patch for SFB (CD45.1−CD90.1+CD4+ T cells) and in LI-LP for Hh-specific (CD45.1+CD90.1−CD4+ T cells) transgenic T cells (n = 4). d, Quantification of RORγt+ Tregs and TH17 cells among total CD4+ T cells in LI-LP of Rorcfl/fl and Foxp3creRorcfl/fl mice (n = 4). e, f, Quantification of MHCII expression on LTi-like ILC3s (n = 9) (e) and eTACs (n = 4) (f) in mLN of H2-Ab1fl/fl and Il22creH2-Ab1fl/fl mice. g, Quantification of RORγt+ Tregs (among Hh-specific CD4+ T cells) and TH17 cells (among Hh-specific CD4+ T cells) were analyzed in LI-LP of H2-Ab1fl/fl and Il22creH2-Ab1fl/fl mice (n = 9). h, Representative flow cytometry plots of the frequency of MHCII expression on ILC3s, DCs in LI-LP of MHCIIneg and MHCIIILC3+ mice (n = 6) as shown in Fig. 3j. i, Frequency of CD44hiCD62Llo ratio among Hh-specific CD4+ T cells were analyzed in mLN and LI-LP for Hh-specific CD4+ T cells as shown in Fig. 3k, l (n = 6). j, k, RORγt+ Tregs in LI-LP of H2-Ab1fl/fl and MHCIIΔILC3 mice (n = 4) were analyzed by flow cytometry. Histogram and MFI of Bim (j). Proportions of Ki-67 positive cells (k). l, Quantification of RORγt+ Tregs among CD4+ T cells in LI-LP of Il2fl/fl and RorccreIl2fl/fl mice (n = 4). m, Quantification of CD25 staining or IL-2 binding in mLN of WT mice (n = 3). Naïve T cells: CD44loCD62Lhi; effector T: CD44hiCD62Llo; TH17 cells: RORγt+FoxP3−; Tregs: FoxP3+; RORγt+Tregs: RORγt+FoxP3+. Data in e, g are pooled from two independent experiments with similar results. Data in b–d, f, i–l are representative of two independent experiments. Data are shown as mean ± s.e.m., statistics shown in m are obtained by one-way ANOVA with Tukey’s multiple comparisons test, statistics shown in b–g, i–l are obtained by unpaired Student’s t-test (two-tailed).

a, Representative flow cytometry plot of the frequency of Itgav expression on LTi-like ILC3s and eTACs in mLN of Itgavfl/fl and RorccreItgavfl/fl mice as shown in Fig. 4d (n = 5). b–d, Quantification of ITGAV on CD4+ T cells (b), LTi-like ILC3s (c) and eTACs (d) in mLN of Itgavfl/fl and Cd4creItgavfl/fl mice (n = 6). e, Quantification of RORγt+ Tregs in mLN of Itgavfl/fl and Cd4creItgavfl/fl mice (n = 6). f, H. Hepaticus (Hh)-specific CD4+ T cells were transferred to Itgavfl/fl and RorccreItgavfl/fl mice colonized with H. hepaticus 14 days before experiment related to Fig. 4f. Data in b–e are representative of two independent experiments. Data are shown as mean ± s.e.m., statistics shown in b–e are obtained by unpaired Student’s t-test (two-tailed).

a, Gating strategy to sort ILCs and T cells from small intestine of the IBD patient for scRNA-seq in Fig. 5. b, Gating strategy to analyse ILC3s and RORγt+ Tregs by flow cytometry as shown in Fig. 5.

a, Violin plot of CD3E expression among clusters of scRNA-seq data as shown in Fig. 5a. b, Bar graph showing the composition of non-T lymphocytes as indicated in Fig. 5a in non-inflamed tissue (NI) versus inflamed tissue (Infla). c, Bar graph showing the composition of ILC3 lymphocytes in non-inflamed tissue (NI) versus inflamed tissue (Infla) from human IBD samples as published44. d, e, A dot plot showing the mean expression (colour) of indicated genes in ILC3 cluster (d) and Treg cluster (e) in non-inflamed versus inflamed tissue from human IBD samples as published44. f, Correlation analyses between the RORγt+ Tregs (RORγt+Helios− among CD4+ T cells) and TH17 cells (RORγt+FoxP3− among CD4+ T cells) in the cohort of CD patients as in Fig. 5d, i, j. g, h, Quantification of frequency of ILC3s among CD127+CD117+ subset (g) and RORγt+ Tregs among total Tregs (h) in a second independent cohort of individuals. Healthy donor n = 15, Crohn’s disease (CD) patients n = 15. i, Correlation analyses between the ILC3 (ILC3 among CD127+CD117+ subset) and RORγt+ Tregs (RORγt+ Helios− among FoxP3+ Tregs) in a second independent cohort of human samples as in (g, h). j, LTi-like ILC3s are necessary and sufficient in selecting for the differentiation fate of microbiota specific RORγt+ Tregs, and selecting against TH17 cells, via antigen presentation with contributions from integrin αv and gradients of competition for IL-2. This collectively enforces immunologic tolerance to microbiota and maintains intestinal homeostasis. Data in g, h are shown as means ± s.e.m., statistics shown in g, h are performed using Mann–Whitney U-test (unpaired), correlative analyses in f, i are compared by Pearson’s rank correlation coefficient (R2). Statistics are calculated by two-tailed test.

List of genes that are differentially expressed between RORγt+ eTACs and LTi-like ILC3s in the mouse mLN.

List of genes that are differentially expressed between RORγt+ Treg cells and RORγt- Treg cells in the human intestine.

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

Springer Nature or its licensor 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.

Lyu, M., Suzuki, H., Kang, L. et al. ILC3s select microbiota-specific regulatory T cells to establish tolerance in the gut. Nature (2022). https://doi.org/10.1038/s41586-022-05141-x

DOI: https://doi.org/10.1038/s41586-022-05141-x

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

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.

Nature (Nature) ISSN 1476-4687 (online) ISSN 0028-0836 (print)

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