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.

  • Letter
  • Published:

Tuft-cell-derived IL-25 regulates an intestinal ILC2–epithelial response circuit

Abstract

Parasitic helminths and allergens induce a type 2 immune response leading to profound changes in tissue physiology, including hyperplasia of mucus-secreting goblet cells1 and smooth muscle hypercontractility2. This response, known as ‘weep and sweep’, requires interleukin (IL)-13 production by tissue-resident group 2 innate lymphoid cells (ILC2s) and recruited type 2 helper T cells (TH2 cells)3. Experiments in mice and humans have demonstrated requirements for the epithelial cytokines IL-33, thymic stromal lymphopoietin (TSLP) and IL-25 in the activation of ILC2s4,5,6,7,8,9,10,11, but the sources and regulation of these signals remain poorly defined. In the small intestine, the epithelium consists of at least five distinct cellular lineages12, including the tuft cell, whose function is unclear. Here we show that tuft cells constitutively express IL-25 to sustain ILC2 homeostasis in the resting lamina propria in mice. After helminth infection, tuft-cell-derived IL-25 further activates ILC2s to secrete IL-13, which acts on epithelial crypt progenitors to promote differentiation of tuft and goblet cells, leading to increased frequencies of both. Tuft cells, ILC2s and epithelial progenitors therefore comprise a response circuit that mediates epithelial remodelling associated with type 2 immunity in the small intestine, and perhaps at other mucosal barriers populated by these cells.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Intestinal tuft cells constitutively express Il25.
Figure 2: Worm infection induces IL-13-dependent tuft cell hyperplasia.
Figure 3: IL-13 signalling in epithelial progenitors gives rise to tuft cell hyperplasia.
Figure 4: Tuft cells regulate intestinal physiology through an ILC2–epithelium response circuit.

Similar content being viewed by others

References

  1. Miller, H. R. & Nawa, Y. Immune regulation of intestinal goblet cell differentiation. Specific induction of nonspecific protection against helminths? Nouv. Rev. Fr. Hematol. 21, 31–45 (1979)

    CAS  PubMed  Google Scholar 

  2. Castro, G. A., Badial-Aceves, F., Smith, J. W., Dudrick, S. J. & Weisbrodt, N. W. Altered small bowel propulsion associated with parasitism. Gastroenterology 71, 620–625 (1976)

    Article  CAS  Google Scholar 

  3. Grencis, R. K. Immunity to helminths: resistance, regulation, and susceptibility to gastrointestinal nematodes. Annu. Rev. Immunol. 33, 201–225 (2015)

    Article  CAS  Google Scholar 

  4. Price, A. E. et al. Systemically dispersed innate IL-13-expressing cells in type 2 immunity. Proc. Natl Acad. Sci. USA 107, 11489–11494 (2010)

    Article  CAS  ADS  Google Scholar 

  5. Neill, D. R. et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature 464, 1367–1370 (2010)

    Article  CAS  ADS  Google Scholar 

  6. Fallon, P. G. et al. Identification of an interleukin (IL)-25-dependent cell population that provides IL-4, IL-5, and IL-13 at the onset of helminth expulsion. J. Exp. Med. 203, 1105–1116 (2006)

    Article  CAS  Google Scholar 

  7. Kang, Z. et al. Epithelial cell-specific Act1 adaptor mediates interleukin-25-dependent helminth expulsion through expansion of Linc-Kit+ innate cell population. Immunity 36, 821–833 (2012)

    Article  CAS  Google Scholar 

  8. Zhao, A. et al. Critical role of IL-25 in nematode infection-induced alterations in intestinal function. J. Immunol. 185, 6921–6929 (2010)

    Article  CAS  Google Scholar 

  9. Van Dyken, S. J. et al. Chitin activates parallel immune modules that direct distinct inflammatory responses via innate lymphoid type 2 and γδ T cells. Immunity 40, 414–424 (2014)

    Article  CAS  Google Scholar 

  10. Owyang, A. M. et al. Interleukin 25 regulates type 2 cytokine-dependent immunity and limits chronic inflammation in the gastrointestinal tract. J. Exp. Med. 203, 843–849 (2006)

    Article  Google Scholar 

  11. Angkasekwinai, P. et al. Interleukin 25 promotes the initiation of proallergic type 2 responses. J. Exp. Med. 204, 1509–1517 (2007)

    Article  CAS  Google Scholar 

  12. Clevers, H. The intestinal crypt, a prototype stem cell compartment. Cell 154, 274–284 (2013)

    Article  CAS  Google Scholar 

  13. Bjerknes, M. et al. Origin of the brush cell lineage in the mouse intestinal epithelium. Dev. Biol. 362, 194–218 (2012)

    Article  CAS  Google Scholar 

  14. Gerbe, F., Legraverend, C. & Jay, P. The intestinal epithelium tuft cells: specification and function. Cell. Mol. Life Sci. 69, 2907–2917 (2012)

    Article  CAS  Google Scholar 

  15. Bezençon, C. et al. Murine intestinal cells expressing Trpm5 are mostly brush cells and express markers of neuronal and inflammatory cells. J. Comp. Neurol. 509, 514–525 (2008)

    Article  Google Scholar 

  16. McKenzie, G. J., Bancroft, A., Grencis, R. K. & McKenzie, A. N. J. A distinct role for interleukin-13 in Th2-cell-mediated immune responses. Curr. Biol. 8, 339–342 (1998)

    Article  CAS  Google Scholar 

  17. Fort, M. M. et al. IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated pathologies in vivo. Immunity 15, 985–995 (2001)

    Article  CAS  Google Scholar 

  18. Hurst, S. D. et al. New IL-17 family members promote Th1 or Th2 responses in the lung: in vivo function of the novel cytokine IL-25. J. Immunol. 169, 443–453 (2002)

    Article  CAS  Google Scholar 

  19. Liang, H.-E. et al. Divergent expression patterns of IL-4 and IL-13 define unique functions in allergic immunity. Nature Immunol. 13, 58–66 (2012)

    Article  CAS  Google Scholar 

  20. Voehringer, D., Reese, T. A., Huang, X., Shinkai, K. & Locksley, R. M. Type 2 immunity is controlled by IL-4/IL-13 expression in hematopoietic non-eosinophil cells of the innate immune system. J. Exp. Med. 203, 1435–1446 (2006)

    Article  CAS  Google Scholar 

  21. Oliphant, C. J. et al. MHCII-mediated dialog between group 2 innate lymphoid cells and CD4+ T cells potentiates type 2 immunity and promotes parasitic helminth expulsion. Immunity 41, 283–295 (2014)

    Article  CAS  Google Scholar 

  22. Muñoz, J. et al. The Lgr5 intestinal stem cell signature: robust expression of proposed quiescent ‘+4’ cell markers. EMBO J. 31, 3079–3091 (2012)

    Article  Google Scholar 

  23. Reinecker, H. C. & Podolsky, D. K. Human intestinal epithelial cells express functional cytokine receptors sharing the common gamma c chain of the interleukin 2 receptor. Proc. Natl Acad. Sci. USA 92, 8353–8357 (1995)

    Article  CAS  ADS  Google Scholar 

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

    Article  CAS  ADS  Google Scholar 

  25. Deckmann, K. et al. Bitter triggers acetylcholine release from polymodal urethral chemosensory cells and bladder reflexes. Proc. Natl Acad. Sci. USA 111, 8287–8292 (2014)

    Article  CAS  ADS  Google Scholar 

  26. Krasteva, G. et al. Cholinergic chemosensory cells in the trachea regulate breathing. Proc. Natl Acad. Sci. USA 108, 9478–9483 (2011)

    Article  CAS  ADS  Google Scholar 

  27. Saunders, C. J., Christensen, M., Finger, T. E. & Tizzano, M. Cholinergic neurotransmission links solitary chemosensory cells to nasal inflammation. Proc. Natl Acad. Sci. USA 111, 6075–6080 (2014)

    Article  CAS  ADS  Google Scholar 

  28. Lee, R. J. et al. Bitter and sweet taste receptors regulate human upper respiratory innate immunity. J. Clin. Invest. 124, 1393–1405 (2014)

    Article  CAS  Google Scholar 

  29. Ballantyne, S. J. et al. Blocking IL-25 prevents airway hyperresponsiveness in allergic asthma. J. Allergy Clin. Immunol. 120, 1324–1331 (2007)

    Article  CAS  Google Scholar 

  30. Han, H., Thelen, T. D., Comeau, M. R. & Ziegler, S. F. Thymic stromal lymphopoietin-mediated epicutaneous inflammation promotes acute diarrhea and anaphylaxis. J. Clin. Invest. 124, 5442–5452 (2014)

    Article  Google Scholar 

  31. Sato, T. & Clevers, H. Primary mouse small intestinal epithelial cell cultures. Methods Mol. Biol. 945, 319–328 (2013)

    Article  Google Scholar 

Download references

Acknowledgements

We thank M. Consengco, R. Noyes, and Z. Wang for technical expertise, Y. Nusse for mice, members of the Locksley laboratory for helpful discussions, and R. Vance, M. Fontana, M. Anderson, and O. Klein for comments on the manuscript. This work was supported by the National Institutes of Health (AI026918, AI030663, HL107202), a Diabetes Endocrinology Research Center grant (DK063720), the Howard Hughes Medical Institute (HHMI), and the Sandler Asthma Basic Research Center at the University of California, San Francisco. J.v.M. is an HHMI Fellow of the Damon Runyon Cancer Research Foundation (DRG-2162-13).

Author information

Authors and Affiliations

Authors

Contributions

J.v.M. conceived the study, designed and performed experiments, analysed data, and wrote the paper with R.M.L. M.J. performed experiments. H.-E.L. cloned the Flare25 reporter cassette, performed the Flare25 embryonic stem cell work, and assisted with additional experiments. R.M.L. directed the study and wrote the paper with J.v.M.

Corresponding author

Correspondence to Richard M. Locksley.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Flare25 mouse and Vil1-cre-mediated Il25 deletion.

a, Gene-targeting strategy for the flox and reporter of Il25 (Flare25) mouse. b, PCR of genomic DNA isolated from the tail (lane 1, 2) or cells sorted from the small intestine (lane 3, 4) of indicated mice. c, Quantitative RT–PCR for Il25 on cDNA from EPCAM+ cells sorted from the small intestine of indicated mice. b, c, Data are representative of two experiments (n = 2). Frt, target site for FLIPASE recombinase; IRES, internal ribosomal entry site; loxP, target site for Cre recombinase; pA, bovine growth hormone poly(A) tail; tdRFP, tandem-dimer red fluorescent protein; UTR, untranslated region. For gel source data (b) see Supplementary Fig. 1.

Extended Data Figure 2 Il25 expression in epithelial surfaces.

a, b, Indicated tissues of Il25F25/F25 (a) and wild-type control (b) mice stained by immunohistochemistry for RFP (red), EPCAM (green), and DAPI (blue). Some data from Fig. 1a are repeated here to allow complete comparison. Scale bars, 50 μm. Images are representative of at least three independent experiments. n = 3.

Extended Data Figure 3 Flow cytometry gating strategies and organoid culture.

a, Flow cytometric analysis of indicated tissues in Il25F25/F25 and wild-type mice. b, Flow cytometric analysis of small intestine epithelial cells of Il25F25/F25 mice before and after fluorescence-activated cell sorting (FACS) into RFP+EPCAM+ and RFPEPCAM+ pools for analysis by quantitative RT–PCR. ce, Representative flow cytometric analysis of small-intestine-derived organoids from Il25F25/F25 (ce) and wild-type (c) mice cultured with or without recombinant protein (20 ng ml−1), as indicated (ce) or Notch signalling inhibitor DAPT (25 μM) (e). Single-cell suspensions of the organoids were stained for EPCAM (ce) and DCLK1 (d), and gated to quantify tuft cell (RFP+EPCAM+ or DCLK1+EPCAM+) frequency. f, Quantification of two technical replicates from experiment shown in e. d.p.i., days post-N. brasiliensis infection. Data in f are technical replicates. Data are representative of three (a, b, d) or two (c, e, f) independent experiments. In ad, n = 3; in e, f, n = 2. Error bars represent mean ± s.e.m.

Extended Data Figure 4 Il25 is expressed constitutively in tuft cells.

ac, Jejunum (a, b) or trachea (c) of Il25F25/F25 (a, c) and wild-type control (b) mice stained by immunohistochemistry for RFP (red), indicated lineage markers (green), and DAPI (blue). Scale bars, 50 μm. Images are representative of one (c) or two (a, b) independent experiments. n = 2.

Extended Data Figure 5 Tuft cells are not a major source of intestinal TSLP or IL-33.

a, Jejunum of Il25F25/F25 mice stained for RFP (red), IL-33 (green), and DAPI (blue). bd, Quantitative RT–PCR on indicated (b, c) or RFP+EPCAM+ (d) cells sorted from untreated (b, c) mice or mice treated as indicated (d). RNA isolated from whole lung 8 days post-N. brasiliensis infection is used as a positive control for Tslp expression in c. Expression of Tslp in sorted Tslp-expressing cells of the lung would probably be higher. Scale bars, 50 μm. Data are representative of two independent experiments. In a, n = 3; in bd, n = 2.

Extended Data Figure 6 N. brasiliensis induces tuft cell hyperplasia throughout the small intestine but not in stomach and colon.

a, b, Indicated tissues of Il25F25/F25 (a) and wild-type control (b) mice treated as indicated and stained by immunohistochemistry for RFP (a) or DCLK1 (b) (red), EPCAM (green), and DAPI (blue). d.p.i., days post-N. brasiliensis infection. Scale bars, 50 μm. Data are representative of two (stomach and colon) or at least three (all others) independent experiments. In a, stomach and colon: n = 2; all others: n > 5.

Extended Data Figure 7 Il25 is expressed only in tuft cells during worm infection and H. polygyrus infection also induces tuft cell hyperplasia.

a, b, Jejunum of Il25F25/F25 (a) and wild-type control (b) mice infected for 7 days with N. brasiliensis stained by immunohistochemistry for RFP (red), indicated lineage markers (green), and DAPI (blue). c, Jejunum of indicated mice left untreated or infected 14 days with H. polygyrus and stained by immunohistochemistry for DAPI (blue), EPCAM (green) and DCLK1 (red). Scale bars, 50 μm. d.p.i., days post-H. polygyrus infection. Images are representative of one (c) or two (a, b) independent experiments. In a, b, n = 2; in c, n = 1 (uninfected) or n = 2 (infected).

Extended Data Figure 8 Absence of Paneth and CHGA+ cell hyperplasia after N. brasiliensis infection and model of ILC2–epithelial signalling circuit.

a, b, Jejunum of indicated mice stained for DAPI (blue) and LYZ1/2 (a) or CHGA (b) (green). c, Quantification of CHGA+ cells from imaging in (b). d, During homeostasis, rare epithelial tuft cells of the small intestine constitutively express Il25, which maintains low levels of IL-13 production in lamina propria ILC2s. IL-13 in turn signals uncommitted epithelial progenitors to promote emergence of tuft and goblet cells. In the absence of infection, this feed-forward ILC2–epithelial circuit is restrained by as yet unknown mechanisms. After N. brasiliensis (N.b.) infection, a helminth-derived signal or a change in host physiology activates the ILC2–epithelial circuit leading to tuft and goblet cell hyperplasia and enhanced IL-13 production by ILC2s. Adaptive TH2 cells probably also provide IL-13 and/or support ILC2 activation, especially when infection or inflammation lasts more than a week. Recombinant proteins are sufficient to induce tuft cell hyperplasia, either by inducing IL-13 production in lymphoid cells (IL-25 or IL-33) or by directly binding epithelial progenitors (IL-4). Scale bars, 50 μm. d.p.i., days post-N. brasiliensis infection. Data in c are biological replicates. Data are representative of two (a) or three (b) independent experiments or pooled from multiple experiments (c). In a, n = 2; in b, c, n is as shown in c. Error bars represent mean ± s.e.m.

Extended Data Figure 9 IL-13 production by lamina propria ILC2 and CD4+ cells.

a, b, Lamina propria cells from Il25−/−;Il13Smart/+, Il13Smart/+, and wild-type control mice analysed by flow cytometry and gated on ILC2 (a, LinCD45+GATA3+) or CD45+CD4+ (b) cells. IL-13 secretion was quantified by measuring surface expression of human CD4, which is expressed from the Il13 locus in Il13Smart reporter mice. c, Frequency of lamina propria CD4+ cells as a percentage of total CD45+ cells as assessed by flow cytometry. d.p.i., days post-N. brasiliensis infection. Data in b, c are biological replicates. Data are representative of at least three (a) independent experiments, or pooled from multiple experiments (b, c). In a, n = 5; in b, c, n is as shown. Error bars represent mean ± s.e.m.

Extended Data Table 1 Antibodies and quantitative RT–PCR primers used in this study

Supplementary information

Supplementary Figure 1

This file contains the gel source data. (PDF 123 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

von Moltke, J., Ji, M., Liang, HE. et al. Tuft-cell-derived IL-25 regulates an intestinal ILC2–epithelial response circuit. Nature 529, 221–225 (2016). https://doi.org/10.1038/nature16161

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature16161

This article is cited by

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