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:

The neuropeptide VIP confers anticipatory mucosal immunity by regulating ILC3 activity

A Correction to this article was published on 30 January 2020

Abstract

Group 3 innate lymphoid cell (ILC3)-mediated production of the cytokine interleukin-22 (IL-22) is critical for the maintenance of immune homeostasis in the gastrointestinal tract. Here, we find that the function of ILC3s is not constant across the day, but instead oscillates between active phases and resting phases. Coordinate responsiveness of ILC3s in the intestine depended on the food-induced expression of the neuropeptide vasoactive intestinal peptide (VIP). Intestinal ILC3s had high expression of the G protein-coupled receptor vasoactive intestinal peptide receptor 2 (VIPR2), and activation by VIP markedly enhanced the production of IL-22 and the barrier function of the epithelium. Conversely, deficiency in signaling through VIPR2 led to impaired production of IL-22 by ILC3s and increased susceptibility to inflammation-induced gut injury. Thus, intrinsic cellular rhythms acted in synergy with the cyclic patterns of food intake to drive the production of IL-22 and synchronize protection of the intestinal epithelium through a VIP–VIPR2 pathway in ILC3s.

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: ILC2 and ILC3 activities oscillate during the active and resting phase at steady state in wild-type mice.
Fig. 2: The molecular clock only partially regulates circadian expression of cytokine secretion.
Fig. 3: Food intake regulates the cytokine secretion of enteric ILC2s and ILC3s.
Fig. 4: Intestinal ILC3 subsets express VIPR2 and are located close to VIP-expressing neurons.
Fig. 5: VIP directly regulates IL-22 production by ILC3s.
Fig. 6: VIPR2 signaling regulates IL-22 secretion from ILC3s in vivo.
Fig. 7: VIPR2 signaling is crucial for the regulation of gut integrity in vivo.
Fig. 8: VIPR2/ ILC3s provide protection from DSS-induced inflammation.

Similar content being viewed by others

Data availability

Single-cell RNA–Seq profiling data that support the findings of this study have been deposited in the Gene Expression Omnibus repository with the accession code GSE132273.

References

  1. Sender, R., Fuchs, S. & Milo, R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell 164, 337–340 (2016).

    CAS  PubMed  Google Scholar 

  2. Scheiermann, C., Kunisaki, Y. & Frenette, P. S. Circadian control of the immune system. Nat. Rev. Immunol. 13, 190–198 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Sonnenberg, G. F. et al. Innate lymphoid cells promote anatomical containment of lymphoid-resident commensal bacteria. Science 336, 1321–1325 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Rankin, L. C. et al. Complementarity and redundancy of IL-22-producing innate lymphoid cells. Nat. Immunol. 17, 179–186 (2016).

    CAS  PubMed  Google Scholar 

  5. Mortha, A. et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 343, 1249288 (2014).

    PubMed  PubMed Central  Google Scholar 

  6. Lindemans, C. A. et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature 528, 560–564 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Zenewicz, L. A. et al. Innate and adaptive interleukin-22 protects mice from inflammatory bowel disease. Immunity 29, 947–957 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Ibiza, S. et al. Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence. Nature 535, 440–443 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Hasegawa, M. et al. Interleukin-22 regulates the complement system to promote resistance against pathobionts after pathogen-induced intestinal damage. Immunity 41, 620–632 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  11. Lee, J. S. et al. AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nat. Immunol. 13, 144–151 (2011).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  13. Zelante, T. et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39, 372–385 (2013).

    CAS  PubMed  Google Scholar 

  14. Lamas, B. et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat. Med. 22, 598–605 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Wang, Y., Mumm, J. B., Herbst, R., Kolbeck, R. & Wang, Y. IL-22 increases permeability of intestinal epithelial tight junctions by enhancing claudin-2 expression. J. Immunol. 199, 3316–3325 (2017).

    CAS  PubMed  Google Scholar 

  16. Peters, C. P., Mjosberg, J. M., Bernink, J. H. & Spits, H. Innate lymphoid cells in inflammatory bowel diseases. Immunol. Lett. 172, 124–131 (2016).

    CAS  PubMed  Google Scholar 

  17. Kirchberger, S. et al. Innate lymphoid cells sustain colon cancer through production of interleukin-22 in a mouse model. J. Exp. Med. 210, 917–931 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Huber, S. et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature 491, 259–263 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Gury-BenAri, M. et al. The spectrum and regulatory landscape of intestinal innate lymphoid cells are shaped by the microbiome. Cell 166, 1231–1246.e13 (2016).

    CAS  PubMed  Google Scholar 

  20. Colonna, M. Innate lymphoid cells: diversity, plasticity, and unique functions in immunity. Immunity 48, 1104–1117 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Laffont, S. et al. Androgen signaling negatively controls group 2 innate lymphoid cells. J. Exp. Med. 214, 1581–1592 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Quatrini, L. et al. Host resistance to endotoxic shock requires the neuroendocrine regulation of group 1 innate lymphoid cells. J. Exp. Med. 214, 3531–3541 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Wallrapp, A. et al. The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature 549, 351–356 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Klose, C. S. N. et al. The neuropeptide neuromedin U stimulates innate lymphoid cells and type 2 inflammation. Nature 549, 282–286 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Cardoso, V. et al. Neuronal regulation of type 2 innate lymphoid cells via neuromedin U. Nature 549, 277–281 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Barrow, A. D. & Colonna, M. Innate lymphoid cell sensing of tissue vitality. Curr. Opin. Immunol. 56, 82–93 (2018).

    PubMed  Google Scholar 

  28. Rankin, L. C. et al. The transcription factor T-bet is essential for the development of NKp46+ innate lymphocytes via the Notch pathway. Nat. Immunol. 14, 389–395 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Mukherji, A., Kobiita, A., Ye, T. & Chambon, P. Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs. Cell 153, 812–827 (2013).

    CAS  PubMed  Google Scholar 

  30. Yu, X. et al. TH17 cell differentiation is regulated by the circadian clock. Science 342, 727–730 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Curtis, A. M., Bellet, M. M., Sassone-Corsi, P. & O’Neill, L. A. Circadian clock proteins and immunity. Immunity 40, 178–186 (2014).

    CAS  PubMed  Google Scholar 

  32. Godinho-Silva, C. et al. Light-entrained and brain-tuned circadian circuits regulate ILC3s and gut homeostasis. Nature 574, 254–258 (2019).

    CAS  PubMed  Google Scholar 

  33. Acosta-Rodriguez, V. A., de Groot, M. H. M., Rijo-Ferreira, F., Green, C. B. & Takahashi, J. S. Mice under caloric restriction self-impose a temporal restriction of food intake as revealed by an automated feeder system. Cell Metab. 26, 267–277.e2 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. van der Maaten, L. & Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008).

    Google Scholar 

  35. Watanabe, K., Vanecek, J. & Yamaoka, S. In vitro entrainment of the circadian rhythm of vasopressin-releasing cells in suprachiasmatic nucleus by vasoactive intestinal polypeptide. Brain Res. 877, 361–366 (2000).

    CAS  PubMed  Google Scholar 

  36. Colwell, C. S. et al. Disrupted circadian rhythms in VIP- and PHI-deficient mice. Am. J. Physiol. Regul. Integr. Comp. Physiol 285, R939–R949 (2003).

    CAS  PubMed  Google Scholar 

  37. Harmar, A. J. et al. The VPAC2 receptor is essential for circadian function in the mouse suprachiasmatic nuclei. Cell 109, 497–508 (2002).

    CAS  PubMed  Google Scholar 

  38. Lelievre, V. et al. Gastrointestinal dysfunction in mice with a targeted mutation in the gene encoding vasoactive intestinal polypeptide: a model for the study of intestinal ileus and Hirschsprung’s disease. Peptides 28, 1688–1699 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Schwede, F., Maronde, E., Genieser, H. & Jastorff, B. Cyclic nucleotide analogs as biochemical tools and prospective drugs. Pharmacol. Ther. 87, 199–226 (2000).

    CAS  PubMed  Google Scholar 

  40. Yadav, M., Huang, M. C. & Goetzl, E. J. VPAC1 (vasoactive intestinal peptide (VIP) receptor type 1) G protein-coupled receptor mediation of VIP enhancement of murine experimental colitis. Cell Immunol. 267, 124–132 (2011).

    CAS  PubMed  Google Scholar 

  41. Veiga-Fernandes, H. & Mucida, D. Neuro-immune interactions at barrier surfaces. Cell 165, 801–811 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Veiga-Fernandes, H. & Pachnis, V. Neuroimmune regulation during intestinal development and homeostasis. Nat. Immunol. 18, 116–122 (2017).

    CAS  PubMed  Google Scholar 

  43. Moriyama, S. et al. β2-adrenergic receptor-mediated negative regulation of group 2 innate lymphoid cell responses. Science 359, 1056–1061 (2018).

    CAS  PubMed  Google Scholar 

  44. Gasteiger, G., Fan, X., Dikiy, S., Lee, S. Y. & Rudensky, A. Y. Tissue residency of innate lymphoid cells in lymphoid and nonlymphoid organs. Science 350, 981–985 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Fan, X. & Rudensky, A. Y. Hallmarks of tissue-resident lymphocytes. Cell 164, 1198–1211 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Huang, Y. et al. S1P-dependent interorgan trafficking of group 2 innate lymphoid cells supports host defense. Science 359, 114–119 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Druzd, D. et al. Lymphocyte circadian clocks control lymph node trafficking and adaptive immune responses. Immunity 46, 120–132 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. 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).

    CAS  PubMed  Google Scholar 

  49. Andoh, A., Bamba, S., Fujiyama, Y., Brittan, M. & Wright, N. A. Colonic subepithelial myofibroblasts in mucosal inflammation and repair: contribution of bone marrow-derived stem cells to the gut regenerative response. J. Gastroenterol. 40, 1089–1099 (2005).

    PubMed  Google Scholar 

  50. Sanos, S. L. et al. RORγt and commensal microflora are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nat. Immunol. 10, 83–91 (2009).

    CAS  PubMed  Google Scholar 

  51. Storch, K. F. et al. Intrinsic circadian clock of the mammalian retina: importance for retinal processing of visual information. Cell 130, 730–741 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  53. Goetzl, E. J. et al. Enhanced delayed-type hypersensitivity and diminished immediate-type hypersensitivity in mice lacking the inducible VPAC2 receptor for vasoactive intestinal peptide. Proc. Natl Acad. Sci. USA 98, 13854–13859 (2001).

    CAS  PubMed  Google Scholar 

  54. Eberl, G. et al. An essential function for the nuclear receptor RORγt in the generation of fetal lymphoid tissue inducer cells. Nat. Immunol. 5, 64–73 (2004).

    CAS  PubMed  Google Scholar 

  55. Garcia, S., DiSanto, J. & Stockinger, B. Following the development of a CD4 T cell response in vivo: from activation to memory formation. Immunity 11, 163–171 (1999).

    CAS  PubMed  Google Scholar 

  56. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    CAS  PubMed  Google Scholar 

  57. Liao, Y., Smyth, G. K. & Shi, W. The Subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 41, e108 (2013).

    PubMed  PubMed Central  Google Scholar 

  58. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

  59. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    PubMed  PubMed Central  Google Scholar 

  60. Robinson, M. D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-Seq data. Genome Biol. 11, R25 (2010).

    PubMed  PubMed Central  Google Scholar 

  61. Gagnon-Bartsch, J. A. & Speed, T. P. Using control genes to correct for unwanted variation in microarray data. Biostatistics 13, 539–552 (2012).

    PubMed  PubMed Central  Google Scholar 

  62. Phipson, B., Lee, S., Majewski, I. J., Alexander, W. S. & Smyth, G. K. Robust hyperparameter estimation protects against hypervariable genes and improves power to detect differential expression. Ann. Appl. Stat. 10, 946–963 (2016).

    PubMed  PubMed Central  Google Scholar 

  63. R Development Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2018).

  64. Huber, W. et al. Orchestrating high-throughput genomic analysis with bioconductor. Nat. Methods 12, 115–121 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Lun, A. T., McCarthy, D. J. & Marioni, J. C. A step-by-step workflow for low-level analysis of single-cell RNA-Seq data with Bioconductor. F1000Res 5, 2122 (2016).

    PubMed  PubMed Central  Google Scholar 

  66. Lun, A. T. L. et al. EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 20, 63 (2019).

    PubMed  PubMed Central  Google Scholar 

  67. McCarthy, D. J., Campbell, K. R., Lun, A. T. & Wills, Q. F. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-Seq data in R. Bioinformatics 33, 1179–1186 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Pijuan-Sala, B. et al. A single-cell molecular map of mouse gastrulation and early organogenesis. Nature 566, 490–495 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Li, W., Germain, R. N. & Gerner, M. Y. Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (Ce3D). Proc. Natl Acad. Sci. USA 114, E7321–E7330 (2017).

    CAS  PubMed  Google Scholar 

  70. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    CAS  PubMed  Google Scholar 

  71. Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91–97 (2018).

    CAS  PubMed  Google Scholar 

  72. Vetizou, M. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 350, 1079–1084 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Mielke, L. A. et al. TCF-1 limits the formation of Tc17 cells via repression of the MAF–RORγt axis. J. Exp. Med. 216, 1682–1699 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Camilleri, A. Lin, S. Cree, C. Alvarado and T. Putoczki for expert technical advice and support. We thank S. Wilcox for performing the sequencing, and S. Nutt for critical reading of the manuscript. Financial support for this work was provided by National Health and Medical Research Council (NHMRC) of Australia grants (APP1165443, 1122277 and 1054925 to G.T.B. and C.S.), Cure Cancer and Cancer Australia (APP1163990 to N.J.), The Rebecca L. Cooper Medical Research Foundation (to G.T.B.) and fellowships from the NHMRC (APP1135898 to G.T.B., APP1123000 to C.S. and APP1154970 to G.K.S.). This study was made possible through the Victorian State Government Operational Infrastructure Support Program and the Australian Government NHMRC Independent Research Institute Infrastructure Support Scheme.

Author information

Authors and Affiliations

Authors

Contributions

C.S., K.L., A.L.G., P.H., N.J., J.T., V.C.W. and R.D.S. performed the experiments and data analyses. G.K.S., A.L.G., P.H. and M.E.R. oversaw the bioinformatic analyses. V.C.W., K.R. and L.W. oversaw the imaging analyses. G.T.B. wrote the draft manuscript and coedited it with C.S. and with the help of the other coauthors. G.T.B. and C.S. directed the studies.

Corresponding authors

Correspondence to Cyril Seillet or Gabrielle T. Belz.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Ioana Visan was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Cytokine secretion of ILCs at T4 and T16.

a,b, Frequency of cytokine-producing NK cells, ILC1, ILC2 and ILC3 isolated from the lungs (a) and mesenteric LN (b) at T4 and T16. IFN-γ, TNF-α, IL-5, IL-13, IL-17 and IL-22 production was determined by intracellular cytokine staining in the indicated populations. Individual responses together with mean ± s.e.m. are shown. a,b, n = 7 mice at each time point. Data is shown for one of four similar experiments for the lung analysis and six experiments for the mesenteric LN analysis. a,b, Statistical significance was determined using a two-tailed unpaired Student’s t-test. *P < 0.05; **P < 0.01; NS, not significant.

Extended Data Fig. 2 Enhanced cytokine expression in ILC subsets at T4 and T16.

Geometric mean fluorescence intensity of intracellular IL-5 and IL-22 cytokine production from ILC2 and ILC3, respectively, isolated from the small intestine of naïve C57BL/6 mice at T4 and T16. Shown are individual responses together with mean ± s.e.m. (n = 4 mice per time point) for one of four similar experiments. Statistical significance was determined using a two-tailed unpaired Student’s t-test. *P < 0.05; **P < 0.01.

Extended Data Fig. 3 Cytokine secretion of T lymphocytes at T4 and T16.

a-c, Frequency of indicated cytokine produced by CD4+ T cells and Th17+ cells (CD4+Rorγt+) in the small intestine (a), lungs (b) and mesenteric LN (c) from naïve C57BL/6 mice at T4 and T16. IFN-γ, TNF-α, IL-17 and IL-22 production was determined by intracellular cytokine staining. Individual responses together with mean ± s.e.m. are shown. a-c, n = 7 mice at each time point. Data show one representative of four for the lung analysis, and one of six similar experiments for the small intestine and mesenteric LN analysis. a-c, Statistical significance was determined using a two-tailed unpaired Student’s t-test. *P < 0.05; **P < 0.01; ***P < 0.001; NS, not significant.

Extended Data Fig. 4 Total number of lymphocytes in the small intestine of mixed bone-marrow chimeric mice.

Mixed bone marrow chimeras were generated by reconstituting CD45.1+/+ lethally irradiated recipients with a 1:1 proportion of F1 (CD45.1 × CD45.2, CD45.1+CD45.2+) wild-type and CD45.2+/+ ArntlΔIL−7R bone marrow. Total number of different lymphocyte subsets isolated in the small intestine of mixed bone marrow chimeras are shown. Cells derived from wild-type (CD45.1+CD45.2+) bone marrow are indicated in white and cells from ArntlΔIL−7R (CD45.2+/+) are shown in black. ILC subsets were gated as live lin(B220CD19CD3) CD45.1+CD45.2+ (wild-type) or CD45.2+/+ (ArntlΔIL−7R) NK1.1+NKp46+ NK cells/ILC1, NK1.1NKp46RorγtGata3+ ILC2 and NK1.1NKp46+/− Rorγt+Gata3 ILC3. Data are representative of three independent experiments and show the mean ± s.e.m. for one experiment (n = 6 mice/experiment). Statistical significance was determined using a two-tailed unpaired Student’s t-test. *P < 0.05; **P < 0.01.

Extended Data Fig. 5 Purification of small intestinal ILC3 from food-restricted mice.

cytometric sorting of lin(CD3εTCRβCD19B220Gr1CD11b) CD45+IL-7Rα+CD90+c-kit+ KLRG1 cells using a BD FACSARIA III (BD Biosciences). a, Representative dot plots show the cell surface markers and gating strategy used to discriminate ILC3 from other ILC subsets. b, Representative analyses of ILC3 purity after cell sorting of 26 independent sorts.

Extended Data Fig. 6 Single cell RNA-sequencing of colon ILC3.

a-c, ILC from colon were sorted and single cells were sequenced using 10× Genomics. a, t-Distributed stochastic neighbour embedding (t-SNE) plots show 7,388 cells (dots) coloured by cluster. b, Level of expression of indicated genes within the t-SNE plots. c, Representative differentially expressed genes (x axis) by cluster (y axis). Dot size represents the fraction of cells within the cluster that express each gene. The colour intensity indicates the z-scaled expression of genes in cells within each cluster. d, Confocal image of a frozen section of the colon of a Rorc(γt)GFP/+ mouse stained for neurons (β3-tubulin III, red), VIP (green), ILC3 (RORγt-GFP+CD3ε, white) and T cells (CD3ε+, purple). Image is representative of 2 independent experiments with 2 mice per experiment. Scale bar, 100 μm. e, Confocal images of frozen sections of colon from Rorc(γt)GFP/+ mice at T4 (n = 68) and T16 (n = 53) stained for neurons (β3-tubulin, red) and VIP (green). Violin plots show the minima, 25% percentile, median, 75% percentile, and maxima for neuron axons in the colon from two mice with 6–8 sections per timepoint. Representative of two independent experiments (n = 2 mice per condition). Scale bar, 50 μm. e, Statistical significance was determined using a two-tailed unpaired Student’s t-test. ***P < 0.001.

Extended Data Fig. 7 Characterisation of lymphocytes in Vipr2/mice.

a, Wild-type and Vipr2/ mice were fed ad libitum or fasted for 16 h and constitutive expression of IL-22 from CD4+ T cells from the small intestine was determined by flow cytometry following 4 h in vitro culture. Data show the individual responses together with the mean ± s.e.m. pooled from two independent experiments (n = 4 Wild-type mice per experiment/condition and n = 2 Vipr2/ mice per experiment/condition). b, Enumeration of ILC1, ILC2, total ILC3, CD4+ ILC3, CD4 ILC3, B cells, CD8+ T cells and CD4+ T cells isolated from the small intestine of naïve wild-type and Vipr2−/− mice. Data show the mean ± s.e.m. of one of two similar experiments (n = 8 mice per time point per experiment). a,b, Statistical significance was determined using a two-tailed unpaired Student’s t-test. *P < 0.05; **P < 0.01; NS, not significant.

Extended Data Fig. 8 Untreated Vipr2/and wild-type mice maintain weight at steady-state.

a, Weight changes for naive control wild-type and Vipr2−/− mice during the DSS treatment period shown in Fig. 7a. Data show mean ± s.e.m (n = 5 mice/genotype) percent of initial weight for one of two independent experiments with similar results. b, Representative H&E stained sections of the colon from untreated wild-type and Vipr2/ mice. Scale bar, 200 μm. Images are representative of two independent experiments with similar results. c-e, Quantitative analysis of crypt height (c), epithelium irregularity (d) and inflammatory infiltrate (e) in untreated WT and Vipr2/ mice. Epithelial length was measured in the distal part of the colon. Overall epithelial irregularity and inflammatory infiltrate was scored from six sections over two different slides per mouse (0 = normal, 1 = minor, 2 = moderate, 3 = severe). Mucosal thickness was measured across 12 randomly selected sites points per mouse. Data show mean ± s.e.m (n = 3 mice/genotype) of one of two independent experiments with similar results. c-e, Statistical significance was determined using a two-tailed unpaired Student’s t-test. *P < 0.05; NS, not significant.

Supplementary information

Reporting Summary

Supplementary Video 1

Nervous innervation of the gut.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Seillet, C., Luong, K., Tellier, J. et al. The neuropeptide VIP confers anticipatory mucosal immunity by regulating ILC3 activity. Nat Immunol 21, 168–177 (2020). https://doi.org/10.1038/s41590-019-0567-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0567-y

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