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:

Intestinal epithelial tuft cells initiate type 2 mucosal immunity to helminth parasites

Abstract

Helminth parasitic infections are a major global health and social burden1. The host defence against helminths such as Nippostrongylus brasiliensis is orchestrated by type 2 cell-mediated immunity2. Induction of type 2 cytokines, including interleukins (IL) IL-4 and IL-13, induce goblet cell hyperplasia with mucus production, ultimately resulting in worm expulsion3,4. However, the mechanisms underlying the initiation of type 2 responses remain incompletely understood. Here we show that tuft cells, a rare epithelial cell type in the steady-state intestinal epithelium5, are responsible for initiating type 2 responses to parasites by a cytokine-mediated cellular relay. Tuft cells have a Th2-related gene expression signature6 and we demonstrate that they undergo a rapid and extensive IL-4Rα-dependent amplification following infection with helminth parasites, owing to direct differentiation of epithelial crypt progenitor cells. We find that the Pou2f3 gene is essential for tuft cell specification. Pou2f3−/− mice lack intestinal tuft cells and have defective mucosal type 2 responses to helminth infection; goblet cell hyperplasia is abrogated and worm expulsion is compromised. Notably, IL-4Rα signalling is sufficient to induce expansion of the tuft cell lineage, and ectopic stimulation of this signalling cascade obviates the need for tuft cells in the epithelial cell remodelling of the intestine. Moreover, tuft cells secrete IL-25, thereby regulating type 2 immune responses. Our data reveal a novel function of intestinal epithelial tuft cells and demonstrate a cellular relay required for initiating mucosal type 2 immunity to helminth infection.

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: Rapid amplification of the tuft cell lineage following infection with Nb.
Figure 2: Absence of tuft cells in the intestinal epithelium of Pou2f3−/− mice.
Figure 3: Impaired type 2 responses in tuft cell-deficient mice.
Figure 4: Tuft cells express IL-25, and rIL-25 is sufficient to initiate type 2 mucosal responses in the absence of tuft cells.

Similar content being viewed by others

References

  1. Hotez, P. J. et al. Helminth infections: the great neglected tropical diseases. J. Clin. Invest. 118, 1311–1321 (2008)

    Article  CAS  Google Scholar 

  2. Allen, J. E. & Maizels, R. M. Diversity and dialogue in immunity to helminths. Nature Rev. Immunol . 11, 375–388 (2011)

    Article  CAS  Google Scholar 

  3. Herbert, D. R. et al. Intestinal epithelial cell secretion of RELM-beta protects against gastrointestinal worm infection. J. Exp. Med. 206, 2947–2957 (2009)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  5. Gerbe, F. et al. Distinct ATOH1 and Neurog3 requirements define tuft cells as a new secretory cell type in the intestinal epithelium. J. Cell Biol. 192, 767–780 (2011)

    Article  CAS  Google Scholar 

  6. 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 

  7. Gerbe, F., Brulin, B., Makrini, L., Legraverend, C. & Jay, P. DCAMKL-1 expression identifies tuft cells rather than stem cells in the adult mouse intestinal epithelium. Gastroenterology 137, 2179–2180 (2009)

    Article  CAS  Google Scholar 

  8. 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 

  9. Buczacki, S. J. et al. Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature 495, 65–69 (2013)

    Article  ADS  CAS  Google Scholar 

  10. van Es, J. H. et al. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nature Cell Biol. 14, 1099–1104 (2012)

    Article  CAS  Google Scholar 

  11. Gierl, M. S., Karoulias, N., Wende, H., Strehle, M. & Birchmeier, C. The zinc-finger factor Insm1 (IA-1) is essential for the development of pancreatic β cells and intestinal endocrine cells. Genes Dev. 20, 2465–2478 (2006)

    Article  CAS  Google Scholar 

  12. Reynolds, L. A., Filbey, K. J. & Maizels, R. M. Immunity to the model intestinal helminth parasite Heligmosomoides polygyrus. Semin. Immunopathol. 34, 829–846 (2012)

    Article  CAS  Google Scholar 

  13. Ishikawa, N., Horii, Y. & Nawa, Y. Immune-mediated alteration of the terminal sugars of goblet cell mucins in the small intestine of Nippostrongylus brasiliensis-infected rats. Immunology 78, 303–307 (1993)

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Watanabe, N., Katakura, K., Kobayashi, A., Okumura, K. & Ovary, Z. Protective immunity and eosinophilia in IgE-deficient SJA/9 mice infected with Nippostrongylus brasiliensis and Trichinella spiralis. Proc. Natl Acad. Sci. USA 85, 4460–4462 (1988)

    Article  ADS  CAS  Google Scholar 

  15. Matsumoto, I., Ohmoto, M., Narukawa, M., Yoshihara, Y. & Abe, K. Skn-1a (Pou2f3) specifies taste receptor cell lineage. Nature Neurosci. 14, 685–687 (2011)

    Article  CAS  Google Scholar 

  16. Ohmoto, M. et al. Pou2f3/Skn-1a is necessary for the generation or differentiation of solitary chemosensory cells in the anterior nasal cavity. Biosci. Biotechnol. Biochem. 77, 2154–2156 (2013)

    Article  CAS  Google Scholar 

  17. Yamaguchi, T. et al. Skn-1a/Pou2f3 is required for the generation of Trpm5-expressing microvillous cells in the mouse main olfactory epithelium. BMC Neurosci . 15, 13 (2014)

    Article  Google Scholar 

  18. Moro, K. et al. Innate production of TH2 cytokines by adipose tissue-associated c-Kit+Sca-1+ lymphoid cells. Nature 463, 540–544 (2010)

    Article  ADS  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  20. Bastide, P. et al. Sox9 regulates cell proliferation and is required for Paneth cell differentiation in the intestinal epithelium. J. Cell Biol. 178, 635–648 (2007)

    Article  CAS  Google Scholar 

  21. Blache, P. et al. SOX9 is an intestine crypt transcription factor, is regulated by the Wnt pathway, and represses the CDX2 and MUC2 genes. J. Cell Biol. 166, 37–47 (2004)

    Article  CAS  Google Scholar 

  22. Mellitzer, G. et al. Loss of enteroendocrine cells in mice alters lipid absorption and glucose homeostasis and impairs postnatal survival. J. Clin. Invest. 120, 1708–1721 (2010)

    Article  CAS  Google Scholar 

  23. Camberis, M., Le Gros, G. & Urban, J. Jr. Animal model of Nippostrongylus brasiliensis and Heligmosomoides polygyrus. Curr. Protoc. Immunol. Ch. 19, Unit 19.12 (2003)

    Google Scholar 

  24. Artis, D. et al. RELMβ/FIZZ2 is a goblet cell-specific immune-effector molecule in the gastrointestinal tract. Proc. Natl Acad. Sci. USA 101, 13596–13600 (2004)

    Article  ADS  CAS  Google Scholar 

  25. Lawrence, R. A., Gray, C. A., Osborne, J. & Maizels, R. M. Nippostrongylus brasiliensis: cytokine responses and nematode expulsion in normal and IL-4-deficient mice. Exp. Parasitol. 84, 65–73 (1996)

    Article  CAS  Google Scholar 

  26. Urban, J. F. Jr et al. IL-13, IL-4Rα, and Stat6 are required for the expulsion of the gastrointestinal nematode parasite Nippostrongylus brasiliensis. Immunity 8, 255–264 (1998)

    Article  CAS  Google Scholar 

  27. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009)

    Article  ADS  CAS  Google Scholar 

  28. 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 

  29. Barner, M., Mohrs, M., Brombacher, F. & Kopf, M. Differences between IL-4Rα-deficient and IL-4-deficient mice reveal a role for IL-13 in the regulation of Th2 responses. Curr. Biol. 8, 669–672 (1998)

    Article  CAS  Google Scholar 

  30. Ohmoto, M., Matsumoto, I., Yasuoka, A., Yoshihara, Y. & Abe, K. Genetic tracing of the gustatory and trigeminal neural pathways originating from T1R3-expressing taste receptor cells and solitary chemoreceptor cells. Mol. Cell. Neurosci. 38, 505–517 (2008)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by Association pour la Recherche contre le Cancer (ARC to N.T. and SL220110603456 to P.J.), Agence Nationale de la Recherche (ANR-09-BLAN-0368-01 to P.J., ANR-PolarAttack to V.D. and ANR-14-CE14-0025-01 to P.J. and N.T.), Institut National du Cancer (INCa 2014-174 to P.J.), a CNRS-NIH International Laboratory grant from the CNRS (LIA-BAGEL) to N.T. and the Welcome Trust (Ref. 106122 to R.M.M.). Part of the work was supported by institutional funds of Monell Chemical Senses Center to I.M.; E.S. and M.B. are supported by Ligue Nationale contre le Cancer, M.P. by the LabEx EpiGenMed and N.T. by Inserm. We are grateful to S. Gailhac and C. Mongellaz for their expertise and assistance in immune cell analyses, S. Cording, J. Di Santo and G. Eberl for their generosity and their expertise on ILCs, G. Petrazzo and R. Guédon for technical input, S. Fre and M. Huygues for technical advice, C Legraverend for her expertise and to M. van de Wetering for reagents. We thank F. Gallardo, the PCEA and the IRD A2 facilities for maintenance of mouse colonies, the Montpellier RIO Imaging (MRI) facility, the Monell Histology and Cellular Localization Core (supported by funding from NIH Core Grant P30DC011735 (to R. F. Margolskee, Monell Chemical Senses Center) for some of the histological analyses and Daniel Fisher for critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

F.G. performed the majority of the experiments. E.S., D.J.S., B.B. and P.C. contributed to mouse studies, M.O. and I.M. to characterization of the Pou2f3-deficient mouse line, E.S., L.G. and M.B. to organoid experiments, V.D., M.P. and V.S.Z. to immune studies and Y.H. to parasite life cycle experiments. P.J. and F.G. conceived the study. P.J., F.G. and R.M.M. designed experiments with contributions from V.D., V.S.Z. and N.T.; P.J. wrote the manuscript with inputs from F.G. and N.T.

Corresponding author

Correspondence to Philippe Jay.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Epithelial differentiation parameters during Nb infection.

a, Graph showing the distribution of Dclk1+ tuft cells in naive and infected mice 4, 5 and 7 days post infection. Cells were counted in the crypt and villus compartments of n = 50 crypt–villus units per mouse with 3 mice per condition. Means of villus/crypt ratio of tuft cell numbers are shown. b, Quantification of the goblet cell hyperplasia in naive and infected mice 4, 5 and 7 days post infection (n = 50 crypt–villus units per mouse; 3 mice per condition). c, Neo-differentiating tuft cells following Nb infection are indistinguishable from the tuft cells found in naive mouse intestinal epithelium as shown with Sox9 and Plcγ2 stainings (n = 3 mice). d, Proliferation status of Pou2f3+ tuft cells in naive and infected mice, shown with co-expression with the Ki67 proliferation marker. Arrows indicate Ki67+ cells located at various positions along the crypt axis. e, Increased proliferation of Pou2f3+ tuft cells during response to Nb infection (n = 3 naive and 3 infected mice). f, Dclk1+ tuft cells and Insm1+ enteroendocrine cells are distinct populations (n = 3 mice). g, Decrease of the Insm1+ enteroendocrine cell population during type 2 responses to Nb infection, concomitant to the expansion of the tuft cell lineage 7 days post infection (n = 3 naive and 3 infected mice). All the histograms show means ± s.d. A two-tailed Student’s t-test with Welch’s correction was used, except for g where the 2 groups displayed comparable variances. All stainings were repeated 3 times.

Extended Data Figure 2 Expansion of the tuft cell lineage is a common adaptation of the intestinal epithelium following infection with helminth parasites.

Tuft cell lineage expansion was assessed by Dclk1 immunohistochemistry in 2 different genetic backgrounds following infection with two different helminths, at the indicated time points. a, b, Naive and Nb-infected BALB/c mice. c, d, Naive and H. polygyrus-infected C57BL/6 and BALB/c mice. e, f, Naive and Nb-infected C57BL/6 and Rag−/− mice. b, d, f, n = 50 crypt–villus units per mouse; 3 mice per condition. Data are shown as means ± s.d. and P values are indicated. A two-tailed Student’s t-test with Welch’s correction was used. Scale bars, 20 μm. All experiments displayed in this figure were repeated 3 times.

Extended Data Figure 3 Pou2f3 deficiency results in the specific absence of tuft cells in the intestinal epithelium.

Characterization of the intestinal epithelium in Pou2f3-deficient mice as compared with wild-type littermate controls (n = 3 mice of each genotype). Left, in situ hybridization showing absence of tuft cells (Pou2f3, Cox1) in Pou2f3−/− mice, whereas enterocytes (Slc5a1), goblet cells (Muc2), Paneth cells (Defcr6) and enteroendocrine cells (glucagon, Gip) are unaffected. Right, representative pictures of the in situ hybridization (Olfm4) and immunohistochemistry experiments underlying the quantitative analysis provided in Fig. 2c showing that the stem cells (Olfm4), proliferative compartment (Ki67), and differentiated cell types: enterocytes (alkaline phosphatase), Paneth (UEA1), enteroendocrine (Insm1) and goblet (PAS staining) cells populations are unaffected in the Pou2f3−/− mice. All panels show representative pictures of experiments replicated 3 times in 3 different mice. Scale bars, 20 μm.

Extended Data Figure 4 Immune cell homeostasis is not altered in Pou2f3−/− tuft-cell-deficient mice.

a, The repartition of immune cells in wild-type and Pou2f3−/− mice was monitored by flow cytometry. The presence of T (CD3+), B (CD19+), CD4+, CD8+, naive (Tn; CD3+CD62L+CD44), central memory (Tcm; CD3+CD62L+CD44+), effector memory (Tem; CD3+CD62LCD44+), regulatory (Treg; CD4+Foxp3+), natural killer (NK; CD3NK1.1+) and myeloid (CD11b+Gr1+) cells was assessed by staining with fluorochrome-tagged antibodies and representative dot plots are shown. The percentages of positively-stained cells are indicated. b, Quantification of the different immune cells in lymph nodes (LN), mesenteric lymph nodes (mLN) and spleens (SP) of wild-type and Pou2f3−/− mice are presented. Data are means ± s.d. (n = 3 mice per genotype). c, Total cells in LN, mLN and SP of wild-type and Pou2f3−/− mice are presented as means ± s.d. (n = 3 mice per genotype). d, Immune lineage cells in the lamina propria of wild-type and Pou2f3−/− mice were monitored by flow cytometry after tissue dissociation. The percentage of T cells was assessed within the CD45+ haematopoietic gate, CD4, CD8 and gamma-delta T cells (CD8+TCR-γδ+) within the CD3+ gate and myeloid cells within the CD3 gate, as indicated. Representative dot plots are presented (left). Quantification of immune cells within the lamina propria are shown as means ± s.d. (n = 3 mice per group). No significant differences were detected for all cell types between wild-type and Pou2f3−/− mice (P > 0.05). A two-tailed Student’s t-test was used.

Extended Data Figure 5 Equivalent immune responsiveness of wild-type and Pou2f3−/− lymphocytes.

a, The level of IL-2 and interferon gamma (IFN-γ) production by Pou2f3+/+ and Pou2f3−/− CD4 and CD8 lymph node T cells was monitored directly after ex vivo isolation and representative histograms are presented (left). Quantification of cytokine secreting CD4 and CD8 T cells are presented as means ± s.d. (n = 3 per group; P > 0.05). b, CFSE-loaded T cells were activated with immobilized anti-CD3/anti-CD28 antibodies for 2 days and proliferation was monitored as a function of fluorescence dilution. Representative histograms for CD4 and CD8 T cells are shown. c, IFN-γ production in wild-type and Pou2f3−/− lymphocytes was assessed at day 6 post CD3/CD28 stimulation and representative plots for CD4 and CD8 T cells are presented. d, Splenocytes from wild-type and Pou2f3−/− mice were activated with LPS+IL-4 for 40 h and levels of secreted IgG, IgG2a, IgG2b and IgA were monitored by ELISA. Means ± s.d. are presented. e, Splenocytes were activated as above and levels of TNF-α, IFN-γ, MCP-1, IL-10, IL-6, and IL-12 were monitored by cytometric bead array. Means ± s.d. are presented. A two-tailed Student’s t-test was used.

Extended Data Figure 6 Defective induction of type 2 immunity in Pou2f3−/− mice following helminth infection.

a, Flow cytometry gating strategy for analysis of the innate ILC2 subset is shown. ILC2s were assessed within the CD45+ haematopoietic subset as lineage-CD127+ cells expressing KLRG1, GATA-3, Sca-1 and CD25 cell surface markers. Numbers represent the percentages of boxed cells. The staining strategy was validated using mLN cells from ZAP-70−/− mice as this subset is present at relatively high levels in these immunodeficient mice. b, The presence of ILC2 cells in mLNs of naive Pou2f3+/+ (WT) and Pou2f3−/− (KO) mice was assessed using the gating strategy shown above. Representative data from WT (n = 8) and KO (n = 5) mice are presented. c, Representative plots of ILC2 cells in lamina propria of naive WT (n = 7) and KO (n = 5) mice are shown (top). Quantifications of ILC2 are presented as means ± s.d. d, WT and KO mice were infected with N. brasiliensis and the presence of ILC2 in mLNs was assessed 5 days post infection. Representative plots are shown (n = 6 mice per group). e, Quantification of ILC2 cells in mLN of naive versus infected WT and KO mice. The percentage of ILC2s within the live gate (left) and the absolute numbers of ILC2s (right) are presented. Data are means ± s.d. (n = 5 for WT, n = 8 for KO, n = 6 for both groups of infected mice). **P = 0.01. f, The fold-increase in ILC2 (lineageCD127+KLRG1+GATA-3+) and Th2 (CD3+CD4+Gata-3+) cells in mLN was assessed as a function of infection (n = 6 per group). The mean fold-increase ± s.d. in WT and KO mice is presented. *P = 0.02, ***P = 0.0005. A two-tailed Student’s t-test was used.

Extended Data Figure 7 Signalling via IL-4Rα is required and sufficient to induce goblet and tuft cell hyperplasia.

a, Quantification of goblet (PAS and Retnlβ staining) cells in Pou2f3+/+ and Pou2f3−/− mice infected with Nb (day 7 post infection). In Pou2f3−/− mice, crypt–villus axes from both focally responding regions and the rest of the tissue were counted. b, Quantification of tuft cells (Dclk1 staining) and goblet cell hyperplasia (PAS) in Il4rα+/+ and Il4rα−/− mice. c, Histological analysis showing tuft (Dclk1 staining) and goblet (PAS and Retnlβ staining) cells in Pou2f3+/+ and Pou2f3−/− mice following treatment with a mixture of rIL-4 and rIL-13 for 5 days. n = 3 mice per condition. All panels show representative experiments replicated 3 times. Scale bars, 20 μm. d, Quantitative analysis of the changes in the different cell types of the intestinal epithelium of Pou2f3+/+ and Pou2f3−/− mice following treatment with a mixture of rIL-4 and rIL-13 during 5 days. For a, b, d, n = 50 crypt–villus axes counted in 3 mice per genotype or condition. Data are shown as means ± s.d. and P values are indicated. A two-tailed Student’s t-test with Welch’s correction was used.

Extended Data Figure 8 Signalling via IL-4Rα is sufficient to induce goblet and tuft cell hyperplasia in mouse intestinal organoids.

a, Quantification of Dclk1 expression analysis by qRT–PCR in Pou2f3+/+ and Pou2f3−/− organoids following rIL-4/rIL-13 treatment for 48 h to assess the presence and amplification of tuft cells. Means ± s.d., relative to Gapdh and Hprt, are presented. b, Expansion of the tuft cell lineage in wild-type organoids following rIL-4/rIL-13 administration (48 h) was monitored by Dclk1 staining. c, Expansion of the tuft cell lineage in wild-type organoids following IL-4 or IL-13 administration (48 h) was monitored by Dclk1, Pou2f3 and PAS stainings. Scale bars, 20 μm. d, Retnlβ expression in Pou2f3+/+ and Pou2f3−/− organoids was monitored as a function of rIL-4/rIL-13 treatment (48 h) by RT–PCR and data relative to Gapdh are presented. All panels show representative experiments from 3 independent organoid cultures, replicated 3 times.

Extended Data Figure 9 Validation of the Siglec-F and IL-25 stainings.

a, Control experiment for specificity of the IL-25 immunohistochemistry, in presence (left) or absence (right) of IL-25 primary antibody. b, Immunohistochemistry showing specificity of Siglec-F as a marker for intestinal epithelial tuft cells. All panels show representative experiments from 3 independent mice, replicated 3 times.

Extended Data Table 1 List of the oligonucleotide primer sequences

Supplementary information

Infected Pou2f3 WT mouse

Mp4 video file showing Nb-infected Pou2f3+/+ mouse with efficient type 2 response to the infection and strong and generalized goblet cell hyperplasia. (MP4 24847 kb)

Infected Pou2f3-deficient mouse

Mp4 video file showing Nb-infected Pou2f3-/- mouse with impaired type 2 response. The rare regions with mild hyperplasia are circled. (MP4 36248 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gerbe, F., Sidot, E., Smyth, D. et al. Intestinal epithelial tuft cells initiate type 2 mucosal immunity to helminth parasites. Nature 529, 226–230 (2016). https://doi.org/10.1038/nature16527

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature16527

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