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:

Group 3 innate lymphoid cells produce the growth factor HB-EGF to protect the intestine from TNF-mediated inflammation

Abstract

Tumor necrosis factor (TNF) drives chronic inflammation and cell death in the intestine, and blocking TNF is a therapeutic approach in inflammatory bowel disease (IBD). Despite this knowledge, the pathways that protect the intestine from TNF are incompletely understood. Here we demonstrate that group 3 innate lymphoid cells (ILC3s) protect the intestinal epithelium from TNF-induced cell death. This occurs independent of interleukin-22 (IL-22), and we identify that ILC3s are a dominant source of heparin-binding epidermal growth factor–like growth factor (HB-EGF). ILC3s produce HB-EGF in response to prostaglandin E2 (PGE2) and engagement of the EP2 receptor. Mice lacking ILC3-derived HB-EGF exhibit increased susceptibility to TNF-mediated epithelial cell death and experimental intestinal inflammation. Finally, human ILC3s produce HB-EGF and are reduced from the inflamed intestine. These results define an essential role for ILC3-derived HB-EGF in protecting the intestine from TNF and indicate that disruption of this pathway contributes to IBD.

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: ILC3s protect the intestinal epithelium from TNF-induced cell death.
Fig. 2: ILC3s are a major cellular source of HB-EGF in the intestine.
Fig. 3: ILC3-derived HB-EGF protects the intestinal epithelium from TNF.
Fig. 4: ILC3s produce PGE2 in response to IL-1β.
Fig. 5: PGE2 elicits ILC3-intrinsic HB-EGF production through the EP2 receptor.
Fig. 6: ILC3-derived HB-EGF protects from acute intestinal damage and inflammation.
Fig. 7: HB-EGF–producing ILC3s protect from chronic intestinal inflammation and are altered in IBD.

Similar content being viewed by others

Data availability

scRNA-seq data are available at Gene Expression Omnibus under accession number GSE166266. All data sets generated and/or analyzed during the current study are presented in this published article, the accompanying source data or supplementary information or are available from the corresponding author upon reasonable request. Source data are provided with this paper.

References

  1. Kalliolias, G. D. & Ivashkiv, L. B. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat. Rev. Rheumatol. 12, 49–62 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Tracey, K. J. & Cerami, A. Tumor necrosis factor: a pleiotropic cytokine and therapeutic target. Annu. Rev. Med. 45, 491–503 (1994).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  4. Graham, D. B. & Xavier, R. J. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature 578, 527–539 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Neurath, M. F. Current and emerging therapeutic targets for IBD. Nat. Rev. Gastroenterol. Hepatol. 14, 269–278 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Patankar, J. V. & Becker, C. Cell death in the gut epithelium and implications for chronic inflammation. Nat. Rev. Gastroenterol. Hepatol. 17, 543–556 (2020).

    Article  PubMed  Google Scholar 

  7. Croft, M., Benedict, C. A. & Ware, C. F. Clinical targeting of the TNF and TNFR superfamilies. Nat. Rev. Drug Discov. 12, 147–168 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Artis, D. & Spits, H. The biology of innate lymphoid cells. Nature 517, 293–301 (2015).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. Monticelli, L. A. et al. IL-33 promotes an innate immune pathway of intestinal tissue protection dependent on amphiregulin-EGFR interactions. Proc. Natl Acad. Sci. USA 112, 10762–10767 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Monticelli, L. A. et al. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat. Immunol. 12, 1045–1054 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Sonnenberg, G. F., Monticelli, L. A., Elloso, M. M., Fouser, L. A. & Artis, D. CD4+ lymphoid tissue-inducer cells promote innate. Immun. Gut Immun. 34, 122–134 (2011).

    CAS  Google Scholar 

  14. Ouyang, W. & O’Garra, A. IL-10 family cytokines IL-10 and IL-22: from basic science to clinical translation. Immunity 50, 871–891 (2019).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  17. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  20. Parker, A. et al. Elevated apoptosis impairs epithelial cell turnover and shortens villi in TNF-driven intestinal inflammation. Cell Death Dis. 10, 108 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Piguet, P. F., Vesin, C., Guo, J., Donati, Y. & Barazzone, C. TNF-induced enterocyte apoptosis in mice is mediated by the TNF receptor 1 and does not require p53. Eur. J. Immunol. 28, 3499–3505 (1998).

    Article  CAS  PubMed  Google Scholar 

  22. Van Hauwermeiren, F. et al. TNFR1-induced lethal inflammation is mediated by goblet and Paneth cell dysfunction. Mucosal Immunol. 8, 828–840 (2015).

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  24. Singh, B. et al. EGF receptor ligands: recent advances.F1000Res. 5, F1000 Faculty Rev-2270 (2016).

    PubMed  PubMed Central  Google Scholar 

  25. Chen, J. et al. Expression and function of the epidermal growth factor receptor in physiology and disease. Physiol. Rev. 96, 1025–1069 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Dao, D. T., Anez-Bustillos, L., Adam, R. M., Puder, M. & Bielenberg, D. R. Heparin-binding epidermal growth factor-like growth factor as a critical mediator of tissue repair and regeneration. Am. J. Pathol. 188, 2446–2456 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Singh, B. & Coffey, R. J. Trafficking of epidermal growth factor receptor ligands in polarized epithelial cells. Annu. Rev. Physiol. 76, 275–300 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Iwamoto, R. et al. Heparin-binding EGF-like growth factor and ErbB signaling is essential for heart function. Proc. Natl Acad. Sci. USA 100, 3221–3226 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Michalsky, M. P., Kuhn, A., Mehta, V. & Besner, G. E. Heparin-binding EGF-like growth factor decreases apoptosis in intestinal epithelial cells in vitro. J. Pediatr. Surg. 36, 1130–1135 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Hilliard, V. C., Frey, M. R., Dempsey, P. J., Peek, R. M. Jr. & Polk, D. B. TNF-alpha converting enzyme-mediated ErbB4 transactivation by TNF promotes colonic epithelial cell survival. Am. J. Physiol. Gastrointest. Liver Physiol. 301, G338–G346 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. El-Assal, O. N. & Besner, G. E. HB-EGF enhances restitution after intestinal ischemia/reperfusion via PI3K/Akt and MEK/ERK1/2 activation. Gastroenterology 129, 609–625 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Elenius, K., Paul, S., Allison, G., Sun, J. & Klagsbrun, M. Activation of HER4 by heparin-binding EGF-like growth factor stimulates chemotaxis but not proliferation. EMBO J. 16, 1268–1278 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Higashiyama, S., Abraham, J. A., Miller, J., Fiddes, J. C. & Klagsbrun, M. A heparin-binding growth factor secreted by macrophage-like cells that is related to EGF. Science 251, 936–939 (1991).

    Article  CAS  PubMed  Google Scholar 

  34. Kalinski, P. Regulation of immune responses by prostaglandin E2. J. Immunol. 188, 21–28 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Dennis, E. A. & Norris, P. C. Eicosanoid storm in infection and inflammation. Nat. Rev. Immunol. 15, 511–523 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Perkins, D. J. et al. Autocrine-paracrine prostaglandin E2 signaling restricts TLR4 internalization and TRIF signaling. Nat. Immunol. 19, 1309–1318 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Sugimoto, Y. & Narumiya, S. Prostaglandin E receptors. J. Biol. Chem. 282, 11613–11617 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Johansson, J. U. et al. Suppression of inflammation with conditional deletion of the prostaglandin E2 EP2 receptor in macrophages and brain microglia. J. Neurosci. 33, 16016–16032 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Gunther, C., Neumann, H., Neurath, M. F. & Becker, C. Apoptosis, necrosis and necroptosis: cell death regulation in the intestinal epithelium. Gut 62, 1062–1071 (2013).

    Article  PubMed  CAS  Google Scholar 

  40. Leppkes, M., Roulis, M., Neurath, M. F., Kollias, G. & Becker, C. Pleiotropic functions of TNF-alpha in the regulation of the intestinal epithelial response to inflammation. Int. Immunol. 26, 509–515 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Gunasekera, D. C. et al. The development of colitis in Il10-/- mice is dependent on IL-22. Mucosal Immunol. 13, 493–506 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Powell, N. et al. Interleukin-22 orchestrates a pathological endoplasmic reticulum stress response transcriptional programme in colonic epithelial cells. Gut 69, 578–590 (2020).

    Article  CAS  PubMed  Google Scholar 

  43. Bouskra, D. et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature 456, 507–510 (2008).

    Article  CAS  PubMed  Google Scholar 

  44. Higashiyama, S. & Nanba, D. ADAM-mediated ectodomain shedding of HB-EGF in receptor cross-talk. Biochim. Biophys. Acta 1751, 110–117 (2005).

    Article  CAS  PubMed  Google Scholar 

  45. Takenobu, H., Yamazaki, A., Hirata, M., Umata, T. & Mekada, E. The stress- and inflammatory cytokine-induced ectodomain shedding of heparin-binding epidermal growth factor-like growth factor is mediated by p38 MAPK, distinct from the 12-O-tetradecanoylphorbol-13-acetate- and lysophosphatidic acid-induced signaling cascades. J. Biol. Chem. 278, 17255–17262 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Ongusaha, P. P. et al. HB-EGF is a potent inducer of tumor growth and angiogenesis. Cancer Res. 64, 5283–5290 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Nakanishi, M. & Rosenberg, D. W. Multifaceted roles of PGE2 in inflammation and cancer. Semin. Immunopathol. 35, 123–137 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Miyoshi, H. et al. Prostaglandin E2 promotes intestinal repair through an adaptive cellular response of the epithelium. EMBO J. 36, 5–24 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Patankar, J. V. et al. E-type prostanoid receptor 4 drives resolution of intestinal inflammation by blocking epithelial necroptosis. Nat. Cell Biol. 23, 796–807 (2021).

    Article  CAS  PubMed  Google Scholar 

  50. Kabashima, K. et al. The prostaglandin receptor EP4 suppresses colitis, mucosal damage and CD4 cell activation in the gut. J. Clin. Investig. 109, 883–893 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Erben, U. et al. A guide to histomorphological evaluation of intestinal inflammation in mouse models. Int. J. Clin. Exp. Pathol. 7, 4557–4576 (2014).

    PubMed  PubMed Central  Google Scholar 

  53. Song-Zhao, G. X. & Maloy, K. J. Experimental mouse models of T cell-dependent inflammatory bowel disease. Methods Mol. Biol. 1193, 199–211 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Sonnenberg Laboratory for discussions and critical reading of the manuscript, the Epigenomics Core of Weill Cornell Medicine and R.C. Harris for sharing critical mouse lines. Research in the Sonnenberg Laboratory is supported by the National Institutes of Health (R01AI143842, R01AI123368, R01AI145989, U01AI095608, R21CA249274 and R01AI162936), the National Institute of Allergy and Infectious Diseases Mucosal Immunology Studies Team, the Searle Scholars Program, the American Asthma Foundation Scholar Award, an Investigators in the Pathogenesis of Infectious Disease Award from the Burroughs Wellcome Fund, a Wade F.B. Thompson/Cancer Research Institute CLIP Investigator grant, the Meyer Cancer Center Collaborative Research Initiative, Linda and Glenn Greenberg, the Dalton Family Foundation and the Roberts Institute for Research in IBD. L.Z. and W.Z. are supported by fellowships from the Crohn’s and Colitis Foundation (608975 and 831404, respectively). C.C. is supported by a Sackler Brain and Spine Institute research grant. A.M.J. is supported by the National Institute of Diabetes and Digestive and Kidney Diseases (T32DK116970). G.F.S. is supported by the Cancer Research Institute Lloyd J. Old STAR Program.

Author information

Authors and Affiliations

Authors

Contributions

L.Z. and G.F.S. conceived the project. L.Z. performed most experiments and analyzed the data. W.Z., A.M.J., C.C., B.F., F.T., M.L. and H.Y. helped with experiments and data analyses. G.G.P. performed bioinformatics analyses. E.M., G.E. and K.I.A. provided essential mouse models, scientific advice and expertise. L.Z. and G.F.S. wrote the manuscript, with input from all authors.

Corresponding authors

Correspondence to Lei Zhou or Gregory F. Sonnenberg.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Arthur Kaser and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Zoltan Fehervari was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Additional information

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

Extended data

Extended Data Fig. 1 The role of ILC3s and IL-22 in TNF-induced intestinal epithelial cell death.

a, Experimental design of the assay. b, Mice in (a) were examined for the depletion efficiency of ILC3s in the small intestine. (n = 3). c, CD90.2 levels in ILC2s or ILC3s were determined in IgG control mice. (n = 3 individual mice). d, Mice in (a) were examined for the depletion efficiency of ILC2s in the small intestine. (n = 3). e, Representative immunofluorescence sections and quantifications of TUNEL staining of the small intestine from noted mice 4 hours post recombinant TNF injection (N = 3 mice per group, n = 24 fields quantified per group). f, Mice in (a) were examined for the neutralization efficiency of endogenous IL-22 (n = 3 individual mice). Data are representative of two independent experiments, shown as the means ± S.E.M. Statistics are calculated by unpaired two-tailed Student’s t-test (b–d and f) or one-way ANOVA (e). Scale bars=50 μm.

Source data

Extended Data Fig. 2 Signature gene expression of ILC clusters in the scRNA-seq data set.

Small intestinal ILCs were sort-purified from PBS- or TNF-treated mice and determined by scRNA-Seq. UMAP shows the relative expression of signature genes in all clusters of each treatment condition.

Extended Data Fig. 3 ILC3s express HB-EGF in the large intestine.

a, Flow cytometry plots show HB-EGF expression in large intestine lamina propria (LI-LP) of noted mice as measured by conversion of the fluorescent LacZ substrate fluorescein di-β-d-galactopyranoside (FDG). b, Graph of frequency of HB-EGF+ cells in SI-LP of noted mice as measured by LacZ activity (n = 5 individual mice). c–e, Flow cytometry plots (c), graph of frequency (d) and quantification of cell number (e) of HB-EGF+ cells in LI-LP of noted mice as measured by LacZ activity (n = 4 individual mice). In both small and large intestines, CD4+ T cells were gated on CD45+CD3ε+CD4+, macrophages were gated on CD45+CD11b+CD11c+MHC II+CD64+, dendritic cells were gated on CD45+CD11b+CD11c+MHCII+CD64-, ILC2s were gated on CD45+Lin- CD127+CD90.2+KLRG1+, ILC3s were gated on CD45+Lin-CD127+CD90.2+KLRG1-CD45dim CCR6+. Lineage 1: CD3ε, CD5, CD8α, NK1.1, TCRγδ. Lineage 2: CD11b, CD11c, B220. Data in a-e are representative of two or three independent experiments, shown as the means ± S.E.M. Statistics are calculated by one-way ANOVA.

Source data

Extended Data Fig. 4 HB-EGF protects from TNF-induced intestinal epithelial cell death.

a–c, Cell death was induced in the murine rectum epithelial cell line CMT-93 by TNF and a cIAP inhibitor. Cell death was determined by representative flow cytometric analysis of cleaved-caspase 3 (a) or cleaved-caspase 8 (c) in the presence or absence of recombinant HB-EGF. Cell death was determined by representative flow cytometric analysis of cleaved-caspase 3 in the presence of various inhibitors (b). Data in a and b are representative of three independent experiments, data in c are pooled from three independent experiments, shown as the means ± S.E.M. Statistics are calculated by one-way ANOVA.

Source data

Extended Data Fig. 5 A PGE2-EP2 axis controls HB-EGF expression in ILC3s.

a, qPCR analysis of Hbegf expression in sort-purified ILC3s cultured with or without TNF treatment, relative to Hprt (n = 4 individual mice). b,c, UMAP show the expression of Ptgs2 (b) or Ptges (c) gene in all clusters of each treatment condition. The dotted line indicates CCR6+ ILC3 clusters. d. qPCR analysis of Hbegf expression in sort-purified ILC3s cultured with or without retinoic acid, relative to Hprt (n = 6). e, Bar graph of the frequencies of IL-17A, IL-22, IFN-γ or GM-CSF in ILC3s in the presence or absence of PGE2 stimulation ex vivo (n = 6). f, qPCR examination of Hbegf transcript in sort-purified ILC3s in the presence of various stimulus, relative to Hprt (n = 4 individual mice). g, Flow cytometry plots with graph of the frequencies of HB-EGF+ immune cells in small intestine of RorccreHbegff/- mice with or without EP2 agonist in vivo treatment (n = 3 individual mice of Mock and n = 4 individual mice of EP2 agonist). h, Western blot analysis of pro-HB-EGF in sort-purified ILC2s or ILC3s from small intestine of Rag1−/− mice with or without EP2 agonist in vivo treatment. i. Sex- and age- matched Ptger2f/f and RorccrePtger2f/f mice were examined for the deletion efficiency of EP2 in ILC3s in the small intestines by qPCR, relative to Hprt (n = 6 individual mice). j, Sort-purified ILC3s were stimulated with IL-1β in the presence or absence of EP2 antagonist, Hbegf expression was then examined by qPCR (n = 6 individual mice). Data in a and f–h are representative of two independent experiments, data in d, e, i and j are pooled from two independent experiments, shown as the means ± S.E.M. Statistics are calculated by unpaired (a, g and i) or paired (d and e) two-tailed Student’s t-test or one-way ANOVA (f and j). *, non-specific band.

Source data

Extended Data Fig. 6 ILC3-derived HB-EGF does not impact immune homeostasis in the healthy gut.

a,b, Flow cytometry plots with graph of frequencies and quantification of cell numbers of ILC3s (a) or IL-22+ ILC3s (b) in the large intestine of Hbegff/f and Hbegf△ILC3 mice at steady state (n = 5 individual mice). c, Graph of frequencies and quantification of cell numbers of T cell subsets in the large intestine of Hbegff/f and Hbegf△ILC3 mice at steady state (n = 5 individual mice). Data in a-c are representative of two independent experiments, shown as the means ± S.E.M. Statistics are calculated by unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 7 Loss of T cell–derived HB-EGF does not alter susceptibility to experimental intestinal inflammation.

a–c, Hbegff/f and Hbegf△T cell mice were given 3% DSS for 6 days, disease and recovery were monitored by weight loss (a), colon shortening (b) and H&E staining of the distal colon (c) at day 11 (n = 5 individual mice). Data in a–c are representative of two independent experiments, shown as the means ± S.E.M. Statistics are calculated by unpaired two-tailed Student’s t-test. Scale bars=100 μm.

Source data

Extended Data Fig. 8 Intestinal epithelial cell responses in the absence of ILC3-specific HB-EGF.

a–d, Hbegff/f and Hbegf△ILC3 mice were challenged with 3% DSS for 6 days (n = 6 individual mice). The functional ability of intestinal epithelium was analyzed, including anti-microbial peptides production (a), tight junction proteins expression (b), stem cell composition (c) and proliferative ability (d) (N = 4 individual mice per group, n = 27 crypts quantified per group). e, Schematic model of chronic colitis induction. Data in a–d are representative of two independent experiments, shown as the means ± S.E.M. Statistics are calculated by unpaired two-tailed Student’s t-test. Scale bars=50 μm.

Source data

Extended Data Fig. 9 ILC3s produce HB-EGF to protect the intestine from TNF.

Here we define a novel pathway of tissue protection in the mammalian intestine. Briefly, ILC3s sense intestinal damage or inflammation through IL-1β release, which amplifies ILC3 responses through a PGE2-EP2 axis, subsequently promoting HB-EGF production and protection against TNF-induced cell death in the intestinal epithelium.

Source data

Supplementary information

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 Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

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. 6

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

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, L., Zhou, W., Joseph, A.M. et al. Group 3 innate lymphoid cells produce the growth factor HB-EGF to protect the intestine from TNF-mediated inflammation. Nat Immunol 23, 251–261 (2022). https://doi.org/10.1038/s41590-021-01110-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01110-0

This article is cited by

Search

Quick links

Nature Briefing

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

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