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:

Basophils prime group 2 innate lymphoid cells for neuropeptide-mediated inhibition

Abstract

Type 2 cytokine responses promote parasitic immunity and initiate tissue repair; however, they can also result in immunopathologies when not properly restricted. Although basophilia is recognized as a common feature of type 2 inflammation, the roles basophils play in regulating these responses are unknown. Here, we demonstrate that helminth-induced group 2 innate lymphoid cell (ILC2) responses are exaggerated in the absence of basophils, resulting in increased inflammation and diminished lung function. Additionally, we show that ILC2s from basophil-depleted mice express reduced amounts of the receptor for the neuropeptide neuromedin B (NMB). Critically, NMB stimulation inhibited ILC2 responses from control but not basophil-depleted mice, and basophils were sufficient to directly enhance NMB receptor expression on ILC2s. These studies suggest that basophils prime ILC2s to respond to neuron-derived signals necessary to maintain tissue integrity. Further, these data provide mechanistic insight into the functions of basophils and identify NMB as a potent inhibitor of type 2 inflammation.

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: Basophils regulate helminth-induced inflammation.
Fig. 2: Basophils negatively regulate ILC2 responses.
Fig. 3: Basophils regulate ILC2 responses independently of T cells.
Fig. 4: Basophils promote the expression of NMBR on ILC2s.
Fig. 5: NMB suppresses helminth-induced type 2 cytokine responses.
Fig. 6: NMBR expression is required to limit helminth-induced inflammation.
Fig. 7: NMB inhibits ILC2-mediated anthelmintic immunity.
Fig. 8: Basophils prime ILC2s for negative regulation by NMB.

Similar content being viewed by others

Data availability

Bulk RNA-seq and single-cell RNA-seq data are deposited in the Gene Expression Omnibus under accession code GSE150793. Source data are provided with this paper.

References

  1. Schistosomiasis and soil-transmitted helminthiases: number of people treated in 2015. Wkly. Epidemiol. Rec. 91, 585–595 (2016).

  2. Jourdan, P. M., Lamberton, P., Fenwick, A. & Addiss, D. G. Soil-transmitted helminth infections. Lancet 391, 252–265 (2018).

    Article  PubMed  Google Scholar 

  3. Jia, T.-W., Melville, S., Utzinger, J., King, C. H. & Zhou, X.-N. Soil-transmitted helminth reinfection after drug treatment: a systematic review and meta-analysis. PLoS Negl. Trop. Dis. 6, e1621 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhan, B. et al. Advancing a multivalent ‘pan-anthelmintic’ vaccine against soil-transmitted nematode infections. Expert Rev. Vaccines 13, 321–331 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  6. Harris, N. L. & Loke, P. Recent advances in type-2-cell-mediated immunity: insights from helminth infection. Immunity 47, 1024–1036 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Gause, W. C., Wynn, T. A. & Allen, J. E. Type 2 immunity and wound healing: evolutionary refinement of adaptive immunity by helminths. Nat. Rev. Immunol. 13, 607–614 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gieseck, R. L. III., Wilson, M. S. & Wynn, T. A. Type 2 immunity in tissue repair and fibrosis. Nat. Rev. Immunol. 18, 62–76 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. Marsland, B. J., Kurrer, M., Reissmann, R., Harris, N. L. & Kopf, M. Nippostrongylus brasiliensis infection leads to the development of emphysema associated with the induction of alternatively activated macrophages. Eur. J. Immunol. 38, 479–488 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Sullivan, B. M. & Locksley, R. M. Basophils: a nonredundant contributor to host immunity. Immunity 30, 12–20 (2009).

    Article  CAS  PubMed  Google Scholar 

  11. Voehringer, D. Protective and pathological roles of mast cells and basophils. Nat. Rev. Immunol. 13, 362–375 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Kim, B. S. et al. Basophils promote innate lymphoid cell responses in inflamed skin. J. Immunol. 193, 3717–3725 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Siracusa, M. C., Kim, B. S., Spergel, J. M. & Artis, D. Basophils and allergic inflammation. J. Allergy Clin. Immunol. 132, 789–801 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Motomura, Y. et al. Basophil-derived interleukin-4 controls the function of natural helper cells, a member of ILC2s, in lung inflammation. Immunity 40, 758–771 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. de Kleer, I. M. et al. Perinatal activation of the interleukin-33 pathway promotes type 2 immunity in the developing lung. Immunity 45, 1285–1298 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Cohen, M. et al. Lung single-cell signaling interaction map reveals basophil role in macrophage imprinting. Cell 175, 1031–1044.e18 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Siracusa, M. C. et al. TSLP promotes interleukin-3-independent basophil haematopoiesis and type 2 inflammation. Nature 477, 229–233 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Webb, L. M. et al. The Notch signaling pathway promotes basophil responses during helminth-induced type 2 inflammation. J. Exp. Med. 216, 1268–1279 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Giacomin, P. R. et al. Thymic stromal lymphopoietin-dependent basophils promote Th2 cytokine responses following intestinal helminth infection. J. Immunol. 189, 4371–4378 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Ohnmacht, C. et al. Basophils orchestrate chronic allergic dermatitis and protective immunity against helminths. Immunity 33, 364–374 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Sullivan, B. M. et al. Genetic analysis of basophil function in vivo. Nat. Immunol. 12, 527–535 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Inclan-Rico, J. M. & Siracusa, M. C. First responders: innate immunity to helminths. Trends Parasitol. 34, 861–880 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wallrapp, A. et al. Calcitonin gene-related peptide negatively regulates alarmin-driven type 2 innate lymphoid cell responses. Immunity 51, 709–723.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nagashima, H. et al. Neuropeptide CGRP limits group 2 innate lymphoid cell responses and constrains type 2 inflammation. Immunity 51, 682–695.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ohki-Hamazaki, H. Neuromedin B. Prog. Neurobiol. 62, 297–312 (2000).

    Article  CAS  PubMed  Google Scholar 

  29. Gajjar, S. & Patel, B. M. Neuromedin: an insight into its types, receptors and therapeutic opportunities. Pharmacol. Rep. 69, 438–447 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Noti, M. et al. Thymic stromal lymphopoietin-elicited basophil responses promote eosinophilic esophagitis. Nat. Med. 19, 1005–1013 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Saenz, S. A., Taylor, B. C. & Artis, D. Welcome to the neighborhood: epithelial cell-derived cytokines license innate and adaptive immune responses at mucosal sites. Immunol. Rev. 226, 172–190 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Stegle, O., Teichmann, S. A. & Marioni, J. C. Computational and analytical challenges in single-cell transcriptomics. Nat. Rev. Genet. 16, 133–145 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Joseph, C. et al. Deciphering hematopoietic stem cells in their niches: a critical appraisal of genetic models, lineage tracing, and imaging strategies. Cell Stem Cell 13, 520–533 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Sutherland, T. E. et al. Chitinase-like proteins promote IL-17-mediated neutrophilia in a tradeoff between nematode killing and host damage. Nat. Immunol. 15, 1116–1125 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chen, F. et al. Neutrophils prime a long-lived effector macrophage phenotype that mediates accelerated helminth expulsion. Nat. Immunol. 15, 938–946 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chen, F. et al. An essential role for TH2-type responses in limiting acute tissue damage during experimental helminth infection. Nat. Med. 18, 260–266 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Minutti, C. M. et al. A macrophage-pericyte axis directs tissue restoration via amphiregulin-induced transforming growth factor beta activation. Immunity 50, 645–654.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ugajin, T. et al. FcεRI, but not FcγR, signals induce prostaglandin D2 and E2 production from basophils. Am. J. Pathol. 179, 775–782 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Siracusa, M. C., Comeau, M. R. & Artis, D. New insights into basophil biology: initiators, regulators, and effectors of type 2 inflammation. Ann. N. Y. Acad. Sci. 1217, 166–177 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Monticelli, L. A. et al. Arginase 1 is an innate lymphoid-cell-intrinsic metabolic checkpoint controlling type 2 inflammation. Nat. Immunol. 17, 656–665 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhou, Y. et al. Prostaglandin E2 inhibits group 2 innate lymphoid cell activation and allergic airway inflammation through E-prostanoid 4-cyclic adenosine monophosphate signaling. Front. Immunol. 9, 501 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Shimokawa, C. et al. Mast cells are crucial for induction of group 2 innate lymphoid cells and clearance of helminth infections. Immunity 46, 863–874.e4 (2017).

    Article  CAS  PubMed  Google Scholar 

  43. Roth, R. L. & Levy, D. A. Nippostrongylus brasiliensis: peripheral leukocyte responses and correlation of basophils with blood histamine concentration during infection in rats. Exp. Parasitol. 50, 331–341 (1980).

    Article  CAS  PubMed  Google Scholar 

  44. Ogilvie, B. M., Hesketh, P. M. & Rose, M. E. Nippostrongylus brasiliensis: peripheral blood leucocyte response of rats, with special reference to basophils. Exp. Parasitol. 46, 20–30 (1978).

    Article  CAS  PubMed  Google Scholar 

  45. Loffredo, L. F. et al. Eosinophil accumulation in postnatal lung is specific to the primary septation phase of development. Sci. Rep. 10, 4425 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Walunas, T. L. et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity 1, 405–413 (1994).

    Article  CAS  PubMed  Google Scholar 

  47. Chikuma, S. et al. PD-1-mediated suppression of IL-2 production induces CD8+ T cell anergy in vivo. J. Immunol. 182, 6682–6689 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Klose, C. S. & Artis, D. Neuronal regulation of innate lymphoid cells. Curr. Opin. Immunol. 56, 94–99 (2019).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Flamar, A.-L. et al. Interleukin-33 induces the enzyme tryptophan hydroxylase 1 to promote inflammatory group 2 innate lymphoid cell-mediated immunity. Immunity 52, 606–619.e6 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Han, H. et al. IL-33 promotes gastrointestinal allergy in a TSLP-independent manner. Mucosal Immunol. 11, 394–403 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Wada, T. et al. Selective ablation of basophils in mice reveals their nonredundant role in acquired immunity against ticks. J. Clin. Investig. 120, 2867–2875 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Bouchery, T. et al. The study of host immune responses elicited by the model murine hookworms Nippostrongylus brasiliensis and Heligmosomoides polygyrus. Curr. Protoc. Mouse Biol. 7, 236–286 (2017).

    Article  CAS  PubMed  Google Scholar 

  54. Ohmori, K. et al. IL-3 induces basophil expansion in vivo by directing granulocyte-monocyte progenitors to differentiate into basophil lineage-restricted progenitors in the bone marrow and by increasing the number of basophil/mast cell progenitors in the spleen. J. Immunol. 182, 2835–2841 (2009).

    Article  CAS  PubMed  Google Scholar 

  55. Anderson, K. G. et al. Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc. 9, 209–222 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Jungblut, M., Oeltze, K., Zehnter, I., Hasselmann, D. & Bosio, A. Standardized preparation of single-cell suspensions from mouse lung tissue using the gentleMACS Dissociator. J. Vis. Exp. 1266 (2009).

  57. Ewels, P. A. et al. The nf-core framework for community-curated bioinformatics pipelines. Preprint at Nat. Biotechnol. 38, 276–278 (2020).

    Article  CAS  PubMed  Google Scholar 

  58. 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).

    Article  CAS  PubMed  Google Scholar 

  59. Pertea, M. et al. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 33, 290–295 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Huang, D. W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Center for Immunity and Inflammation for discussions and critical reading, the New Jersey Medical School (NJMS) Flow Cytometry and Immunology Core Laboratory for technical assistance and the NJMS Histology Core and the NJMS Genomics Research Lab for bioinformatics assistance. We thank D. Voehringer for kindly providing the Mcpt8Cre mice and D. Sant’Angelo for providing the OP9-DL1 cell line. This work was supported by the National Institutes of Health (grant nos. RO1 AI123224 and RO1 AI131634 to M.C.S., T32AI125185 to C.B.S. and 3R01AI131634-02W1 to C.M.H.). J.M.I.R. is supported by the Mexican Council of Science & Technology (CVU 536876).

Author information

Authors and Affiliations

Authors

Contributions

J.M.I.R., J.J.P., N.V.P., C.M.H., C.B.S., A.D.L. and M.C.S. designed and performed the research. A.M.B. contributed to experimental design and data analysis, conceptualization and manuscript editing. J.M.I.R. and M.C.S. analyzed the experimental data and wrote the paper.

Corresponding author

Correspondence to Mark C. Siracusa.

Ethics declarations

Competing interests

Mark C. Siracusa is the founder and president of Nemagen Discoveries.

Additional information

Peer review information Jamie D. K. Wilson 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 Basophils limit helminth-induced pulmonary inflammation.

a, Supernatant levels of IL-4, IL-5 and IL-13 from re-stimulated mesenteric lymph nodes (mLNs) isolated from control or basophil-depleted mice. Mucus production was evaluated in control and basophil-depleted mice on day 7 post-Nb infection by (b), periodic acid shiff (PAS) staining and (c), Muc5ac expression in the lungs by real-time PCR. d, Lung pathology was evaluated by H&E-stained sections with individual images digitally tiled together to provide a larger overview. P values were determined by two-tailed Student’s t-tests. a-d, Representative of at least 3 separate experiments with at least 5 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001. (b), Illustrate data pooled from 2 separate experiments.

Source data

Extended Data Fig. 2 Basophil depletion results in elevated ILC2 responses.

a, Lung neutrophils and (b), eosinophils were quantified by flow cytometry on day 7 post-Nb infection in control or baso-dep mice. c, Representative flow cytometric gating strategy to evaluate neutrophils and eosinophils. d, ILC2s in the lung were quantified on day 7 post-Nb infection in control or baso-dep mice. Intracellular cytokine staining for (e, f), IL-5 and IL-13 was performed on lineage negative, CD90+, CD127+ ILC2s in lung on day 7 post-Nb infection and cytokine positive cells were quantified. g, Representative flow cytometric gating strategy to evaluate ILC2 populations. P values were determined by two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001. a-g, Representative of at least 3 separate experiments with at least 5 mice per group.

Source data

Extended Data Fig. 3 Constitutive ablation of basophils is associated with increased ILC2 activation.

a, b, IL-5+ and IL-13+ ILC2s, as well as (c), eosinophils in the BAL and (d-f), lungs were quantified in control and Mcpt8Cre-4get mice that constitutively lack basophils, 7 days post-Nb infection. P values were determined by two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001. a-f, Representative of at least 3 separate experiments with at least 5 mice per group.

Source data

Extended Data Fig. 4 Basophils are sufficient to limit helminth-induced ILC2 responses.

a, ILC2 numbers, (b), ILC2 production of IL-5, and (c), IL-13, as well as (d) eosinophil numbers were quantified in the lung on day 7 post-Nb infection in control mice, baso-dep mice, or baso-dep mice that received adoptive transfers of basophils. e, H&E staining of lung sections on day 7 post-Nb infection with individual images digitally tiled together to provide a larger overview. Mucus production in the lung was evaluated by (f), PAS staining of lung sections and (g), Muc5ac expression. P values were determined by two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001. a-g, Representative of at least 3 separate experiments with at least 4 mice per group.

Source data

Extended Data Fig. 5 Basophils regulate ILC2s independently of adaptive lymphocytes.

Nb-infected Rag2-/- mice were treated with isotype control or the basophil-depleting antibody MAR-1 and (a), ILC2 responses and (b), eosinophilia were determined in the BAL and (c,d), lung on day 7 post-infection. (e), H&E staining of lung sections on day 7 post-Nb infection with individual images digitally tiled together to provide a larger overview. (f), Mucus production in the lung was evaluated by Muc5ac expression. P values were determined by two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001. a-f, Representative of at least 3 separate experiments with at least 2 mice per naive groups and at least 4 mice per infected groups.

Source data

Extended Data Fig. 6 Elevated ILC2 responses are not associated with increased cytokine alarmin expression.

a-c, Expression of cytokine alarmins in the lungs of control and baso-dep mice was determined on day 7 post-Nb infection by real-time PCR. d, e, Numbers of IL-33-GFP+ type 1 and type 2 pneumocytes were evaluated in IL-33-GFP-reporter mice infected with Nb and treated with the basophil-depleting antibody MAR-1. Expression of (f), Il10 and (g), Areg in the lungs of control and baso-dep mice was determined on day 7 post-Nb infection by real-time PCR. Splenic basophils were sort-purified and cultured (O/N) with IL-3 and anti-IgE antibody and supernatant levels of (h), IL-6, (i), amphiregulin (Areg), and (j), IL-10 were evaluated by ELISA. P values were determined by two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001. a-g, Representative of at least 2 separate experiments with at least 2 mice per naive groups and at least 5 mice per infected groups. h-j, Representative of at least 3 separate experiments with at least 5 individual samples of sort-purified basophils from 5 mice per experimental group.

Source data

Extended Data Fig. 7 Single cell RNAseq analysis of lung-resident ILC2s.

a, Uniform Manifold Approximation and Projection (UMAP) plot illustrating defined clusters of cells generated by single cell RNA-sequencing of lung-resident live ILC2 populations (CD45+Lin-CD90+CD127+) sort-purified from control (and basophil-depleted (baso-dep) mice 5 days post-Nb infection. b, Top 10 marker genes expressed by each cluster of ILC2s. c, Single-cell expression of Il5, Il13, Areg, Arg1, Il1rl1, and Il17rb in ILC cell clusters as defined in A. Horizontal bars represent mean normalized expression. P values were determined by Wilcoxon signed rank sum test. *P < 0.05, **P < 0.01, ***P < 0.001.

Source data

Extended Data Fig. 8 Analysis of NMBR expression in the hematopoietic compartment.

a, Heat map illustrating representative genes of interest expressed in control or baso-dep ILC2s. Surface NMBR expression by (b), CD4+ T cells, (c), alveolar macrophages, (d), non-alveolar macrophages, (e), neutrophils, and (f), eosinophils was determined in lung suspensions of naïve and mice infected with Nb 7 days prior. P values were determined by two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001. (b-f), Representative of at least 3 separate experiments with at least 2 mice per naive groups and at least 4 mice per infected groups.

Source data

Extended Data Fig. 9 NMB-NMBR signaling suppresses helminth-induced ILC2 responses.

a, Schematic illustrating targeting strategy and placement of loxP cassettes upstream and downstream of exon 2 of the Nmbr gene. b-d, Type 2 cytokine expression in the lungs of NMBRfl/fl controls and NMBRfl/fl x Vav-iCre+ mice was determined on day 7 post-Nb infection by real-time PCR. e, IL-5+ and (f), IL-13+ ILC2s, as well as (g), eosinophils were quantified in the lungs of NMBRfl/fl x Vav-iCre+ mice 7 days post-Nb. Nb-infected Rag2-/- mice were treated with PBS or rNMB (i.t.) and (h, i), the percentage of IL-5+ and IL-13+ ILC2s were determined in the BAL and (j), the total number of IL-5+ and IL-13+ ILC2s were determined in the lung on day 7 post-infection. k, eosinophils and (l), neutrophils were determined in the lungs of Rag2-/- mice treated with PBS or rNMB on day 7-post infection. P values were determined by two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001. b-l, Representative of 3 separate experiments with at least 3 mice per naive groups and at least 5 mice per infected groups.

Source data

Extended Data Fig. 10 Basophils are required for NMBR-mediated inhibition of ILC2s.

Sort-purified ILC2s were cultured (O/N) with vehicle or rNMB in the presence of IL-2 and IL-7 or IL-2, IL-7, and IL-33. a, b, The percentage of IL-5+ and IL-13+ ILC2s were quantified by intracellular staining. c, d, IL-5 and IL-13 levels in the supernatant were quantified by ELISA. Sort-purified ILC2s were cultured (O/N) alone or with activated basophils. e, Cytokine levels in the supernatant were monitored by ELISA and (f, g), cell proliferation was evaluated by CTV dilution 4 days post-culture. h, Heat map illustrating genes differentially expressed at 2.0-fold or higher between control or NMB-treated ILC2s. i, Heat map illustrating genes not differentially expressed in control or NMB-treated ILC2s. P values were determined by two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001. a-g, Representative of at least 3 separate experiments with at least 5 individual samples of sort-purified ILC2s in each experimental group.

Source data

Supplementary information

Reporting Summary

Supplementary Table

Supplementary Tables 1 and 2.

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 Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

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

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Inclan-Rico, J.M., Ponessa, J.J., Valero-Pacheco, N. et al. Basophils prime group 2 innate lymphoid cells for neuropeptide-mediated inhibition. Nat Immunol 21, 1181–1193 (2020). https://doi.org/10.1038/s41590-020-0753-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0753-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