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:

Type I interferon restricts type 2 immunopathology through the regulation of group 2 innate lymphoid cells

Abstract

Viral respiratory tract infections are the main causative agents of the onset of infection-induced asthma and asthma exacerbations that remain mechanistically unexplained. Here we found that deficiency in signaling via type I interferon receptor led to deregulated activation of group 2 innate lymphoid cells (ILC2 cells) and infection-associated type 2 immunopathology. Type I interferons directly and negatively regulated mouse and human ILC2 cells in a manner dependent on the transcriptional activator ISGF3 that led to altered cytokine production, cell proliferation and increased cell death. In addition, interferon-γ (IFN-γ) and interleukin 27 (IL-27) altered ILC2 function dependent on the transcription factor STAT1. These results demonstrate that type I and type II interferons, together with IL-27, regulate ILC2 cells to restrict type 2 immunopathology.

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

Figure 1: Deficiency in signaling via type I interferon receptor results in increased ILC2 cells and type 2 immunopathology.
Figure 2: Type I interferons restrain the proliferation and cytokine production of mouse ILC2 cells.
Figure 3: Human ILC2 cells are regulated by type I interferons.
Figure 4: Mechanisms of type I interferon–mediated regulation of ILC2 cells.
Figure 5: Stimulation of ILC2 cells with type I interferons reduces proliferation, viability and cytokine production.
Figure 6: Type I interferons restrict ILC2 cells and type 2 immunopathology in vivo.
Figure 7: IL-27 and IFN-γ regulate ILC2 function.
Figure 8: IFN-γ restricts ILC2 cells and type 2 immunopathology in vivo.

Similar content being viewed by others

References

  1. 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 

  2. Hoyler, T. et al. The transcription factor GATA-3 controls cell fate and maintenance of type 2 innate lymphoid cells. Immunity 37, 634–648 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  4. Diefenbach, A., Colonna, M. & Koyasu, S. Development, differentiation, and diversity of innate lymphoid cells. Immunity 41, 354–365 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. McKenzie, A.N., Spits, H. & Eberl, G. Innate lymphoid cells in inflammation and immunity. Immunity 41, 366–374 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. Halim, T.Y. et al. Group 2 innate lymphoid cells are critical for the initiation of adaptive T helper 2 cell-mediated allergic lung inflammation. Immunity 40, 425–435 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Barlow, J.L. & McKenzie, A.N. Type-2 innate lymphoid cells in human allergic disease. Curr. Opin. Allergy Clin. Immunol. 14, 397–403 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lambrecht, B.N. & Hammad, H. The immunology of asthma. Nat. Immunol. 16, 45–56 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Yoo, J.K., Kim, T.S., Hufford, M.M. & Braciale, T.J. Viral infection of the lung: host response and sequelae. J. Allergy Clin. Immunol. 132, 1263–1276, quiz 1277 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hansel, T.T., Johnston, S.L. & Openshaw, P.J. Microbes and mucosal immune responses in asthma. Lancet 381, 861–873 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Edwards, M.R., Bartlett, N.W., Hussell, T., Openshaw, P. & Johnston, S.L. The microbiology of asthma. Nat. Rev. Microbiol. 10, 459–471 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wark, P.A. et al. Asthmatic bronchial epithelial cells have a deficient innate immune response to infection with rhinovirus. J. Exp. Med. 201, 937–947 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Edwards, M.R. et al. Impaired innate interferon induction in severe therapy resistant atopic asthmatic children. Mucosal Immunol. 6, 797–806 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. García-Sastre, A. Induction and evasion of type I interferon responses by influenza viruses. Virus Res. 162, 12–18 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Barik, S. Respiratory syncytial virus mechanisms to interfere with type 1 interferons. Curr. Top. Microbiol. Immunol. 372, 173–191 (2013).

    CAS  PubMed  Google Scholar 

  17. Feng, Q., Langereis, M.A. & van Kuppeveld, F.J. Induction and suppression of innate antiviral responses by picornaviruses. Cytokine Growth Factor Rev. 25, 577–585 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Jain, S. et al. Hospitalized patients with 2009 H1N1 influenza in the United States, April-June 2009. N. Engl. J. Med. 361, 1935–1944 (2009).

    Article  CAS  PubMed  Google Scholar 

  19. Terai, M. et al. Early induction of interleukin-5 and peripheral eosinophilia in acute pneumonia in Japanese children infected by pandemic 2009 influenza A in the Tokyo area. Microbiol. Immunol. 55, 341–346 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Seo, S.U. et al. Type I interferon signaling regulates Ly6Chi monocytes and neutrophils during acute viral pneumonia in mice. PLoS Pathog. 7, e1001304 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chang, Y.J. et al. Innate lymphoid cells mediate influenza-induced airway hyper-reactivity independently of adaptive immunity. Nat. Immunol. 12, 631–638 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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  PubMed  PubMed Central  Google Scholar 

  23. Brickshawana, A., Shapiro, V.S., Kita, H. & Pease, L.R. LineageSca1+c-KitCD25+ cells are IL-33-responsive type 2 innate cells in the mouse bone marrow. J. Immunol. 187, 5795–5804 (2011).

    Article  CAS  PubMed  Google Scholar 

  24. Mjösberg, J. et al. The transcription factor GATA3 is essential for the function of human type 2 innate lymphoid cells. Immunity 37, 649–659 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Trinchieri, G. Type I interferon: friend or foe? J. Exp. Med. 207, 2053–2063 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gupta, S., Jiang, M. & Pernis, A.B. IFN-α activates Stat6 and leads to the formation of Stat2:Stat6 complexes in B cells. J. Immunol. 163, 3834–3841 (1999).

    CAS  PubMed  Google Scholar 

  27. Huber, J.P. & Farrar, J.D. Regulation of effector and memory T-cell functions by type I interferon. Immunology 132, 466–474 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Diefenbach, A. et al. Type 1 interferon (IFNα/β) and type 2 nitric oxide synthase regulate the innate immune response to a protozoan parasite. Immunity 8, 77–87 (1998).

    Article  CAS  PubMed  Google Scholar 

  29. Barlow, J.L. et al. IL-33 is more potent than IL-25 in provoking IL-13-producing nuocytes (type 2 innate lymphoid cells) and airway contraction. J. Allergy Clin. Immunol. 132, 933–941 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Murray, P.J. et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 14–20 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Duffield, J.S., Lupher, M., Thannickal, V.J. & Wynn, T.A. Host responses in tissue repair and fibrosis. Annu. Rev. Pathol. 8, 241–276 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Kumamoto, Y. et al. CD301b+ dermal dendritic cells drive T helper 2 cell-mediated immunity. Immunity 39, 733–743 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Murakami, R. et al. A unique dermal dendritic cell subset that skews the immune response toward Th2. PLoS ONE 8, e73270 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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 

  35. Sweeney, C.M. et al. IL-27 mediates the response to IFN-beta therapy in multiple sclerosis patients by inhibiting Th17 cells. Brain Behav. Immun. 25, 1170–1181 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. García-Sastre, A. et al. The role of interferon in influenza virus tissue tropism. J. Virol. 72, 8550–8558 (1998).

    PubMed  PubMed Central  Google Scholar 

  37. Price, G.E., Gaszewska-Mastarlarz, A. & Moskophidis, D. The role of α/β and γ interferons in development of immunity to influenza A virus in mice. J. Virol. 74, 3996–4003 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Blazejewska, P. et al. Pathogenicity of different PR8 influenza A virus variants in mice is determined by both viral and host factors. Virology 412, 36–45 (2011).

    Article  CAS  PubMed  Google Scholar 

  39. Kernbauer, E., Ding, Y. & Cadwell, K. An enteric virus can replace the beneficial function of commensal bacteria. Nature 516, 94–98 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sciumé, G. et al. Distinct requirements for T-bet in gut innate lymphoid cells. J. Exp. Med. 209, 2331–2338 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Klose, C.S. et al. A T-bet gradient controls the fate and function of CCR6-RORγt+ innate lymphoid cells. Nature 494, 261–265 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Hunter, C.A. & Kastelein, R. Interleukin-27: balancing protective and pathological immunity. Immunity 37, 960–969 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Moro, K. et al. Interferon and IL-27 antagonize the function of group 2 innate lymphoid cells and type 2 innate immune responses. Nat. Immunol. 10.1038/ni.3309 (23 November 2015).

  45. Kumar, A. & Ghosh, B. A single nucleotide polymorphism (A → G) in intron 3 of IFNγ gene is associated with asthma. Genes Immun. 9, 294–301 (2008).

    Article  CAS  PubMed  Google Scholar 

  46. Pinto, L.A. et al. STAT1 gene variations, IgE regulation and atopy. Allergy 62, 1456–1461 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Nakao, F. et al. Association of IFN-γ and IFN regulatory factor 1 polymorphisms with childhood atopic asthma. J. Allergy Clin. Immunol. 107, 499–504 (2001).

    Article  CAS  PubMed  Google Scholar 

  48. Kroegel, C. et al. Interferon-alphacon-1 treatment of three patients with severe glucocorticoid-dependent asthma. Effect on disease control and systemic glucocorticosteroid dose. Respiration 73, 566–570 (2006).

    Article  PubMed  Google Scholar 

  49. Gaush, C.R. & Smith, T.F. Replication and plaque assay of influenza virus in an established line of canine kidney cells. Appl. Microbiol. 16, 588–594 (1968).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Ruest, A., Michaud, S., Deslandes, S. & Frost, E.H. Comparison of the Directigen flu A+B test, the QuickVue influenza test, and clinical case definition to viral culture and reverse transcription-PCR for rapid diagnosis of influenza virus infection. J. Clin. Microbiol. 41, 3487–3493 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ward, C.L. et al. Design and performance testing of quantitative real time PCR assays for influenza A and B viral load measurement. J. Clin. Virol. 29, 179–188 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Flaczyk, A. et al. IL-33 signaling regulates innate and adaptive immunity to Cryptococcus neoformans. J. Immunol. 191, 2503–2513 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Kaufman, G.N. et al. Intravenous immunoglobulin attenuates airway hyperresponsiveness in a murine model of allergic asthma. Clin. Exp. Allergy 41, 718–728 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Maazi, H. et al. ICOS:ICOS-ligand interaction is required for type 2 innate lymphoid cell function, homeostasis, and induction of airway hyperreactivity. Immunity 42, 538–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J.L. Gommerman (University of Toronto) for Ifnar1−/− (Ifnar1tm1Agt) mice; D. Xue and G.A. Kaufman for help with AHR measurements; and B. Charbonneau for technical support. Supported by the Canadian Institutes of Health Research (J.H.F. and C.U.D.; MOP-114972 for the J.H.F. laboratory; postdoctoral fellowship for C.U.D.; MOP-89821 operating funds for S.M.V.), the Canadian Foundation of Innovation (Leaders Opportunity Fund infrastructure grant for the J.H.F. laboratory), the German National Academy of Sciences Leopoldina (C.U.D.), The American Association of Immunologists Careers in Immunology Fellowship Program (J.H.F. and C.U.D.) and Canada Research Chair in Host Responses to Virus Infections (S.M.V.).

Author information

Authors and Affiliations

Authors

Contributions

C.U.D. and J.H.F. designed the conceptual framework of the study, designed experiments and wrote the paper; C.U.D., C.D.A.M. and B.C.M. designed and performed experiments and analyzed data; M.R. and M.S. performed experiments with human ILC2 cells and helped to design and interpret experiments; A.P.M. and I.L.K. provided H. polygyrus, performed infections and helped to design and interpret experiments; J.P. and S.M.V. provided influenza virus and helped to design and interpret experiments; M.M.E. and D.M. provided STAT4-mutant mice; J.-F.G. provided reagents and expertise for experiments with IL-27; S.T.Q. provided STAT1-deficient BM; B.D.M. provided equipment and expertise for AHR measurements; K.L.M. provided IRF9-deficient BM; A.M.G. provided STAT1- and STAT2-deficient BM; and all authors provided input throughout the study and the writing of the manuscript.

Corresponding author

Correspondence to Jörg H Fritz.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Wild-type and Ifnar1−/− mice recover from infection with a low dose of IAV equally well.

(a,b) WT and Ifnar1-/- mice (five age- and sex-matched animals per group) were either mock treated with PBS as a control or infected with 20 PFU of influenza virus. (a) Survival and (b) body weight loss were monitored throughout the course of infection. (c) Pulmonary expression of the non-structural 1 (Ns1) gene of influenza was determined by qRT-PCR 5, 10 and 15 days post infection. The bars represent the mean ± standard deviation of each cohort. Data are representative of two independent experiments; n.d., not detectable. (d-g) WT and Ifnar1-/- mice (three to seven age- and sex-matched animals per group) were either mock treated with PBS or infected intranasally with 20 PFU of influenza virus. Five days post infection lungs were perfused with PBS and the pulmonary expression levels of (d) IL1b, (e) Tnf and (f) Ccl2 were determined by qRT-PCR, and (g) the numbers of inflammatory monocytes were determined by flow cytometry. Data are representative of three independent experiments. The bars represent the mean ± standard deviation of triplicates of each cohort. The asterisks indicate statistically significant differences (*, p ≤ 0.05; **, p ≤ 0.01; ***, p ≤ 0.001). (h) WT and Ifnar1-/- mice (three to seven age- and sex-matched animals per group) were either mock treated with PBS or infected intranasally with 20 PFU of influenza virus. At indicated time points post infection lungs were perfused with PBS, homogenized, and the IL-33 content was determined by ELISA. Data are representative of two independent experiments. The bars represent the mean ± standard deviation of triplicates of each cohort. One-way ANOVA test including the Bonferroni’s multiple comparison was used to analyze statistical significance.

Supplementary Figure 2 IFNAR1 deficiency results in elevated innate and adaptive type 2 immunity upon infection with H. polygyrus.

(a,b) WT and Ifnar1-/- mice (three to five age- and sex-matched animals per group) were either mock treated with PBS as control (co) or infected with H. polygyrus (H. poly., 200 larvae/mouse). Counts of (a) neutrophils, eosinophils and (b) group 2 innate lymphoid cells (ILC2) in the lamina propria of the small intestine and (c) IgG1 and (d) IgE levels in sera of mice were determined five days post-infection (p.i.). Data are representative of two independent experiments. The asterisks indicate statistically significant differences (*, p ≤ 0.05; **, p ≤ 0.01).

Supplementary Figure 3 Isolation and ex vivo expansion of mouse BM-derived ILC2 cells.

(a) Schematic representation of isolation and ex vivo expansion of murine bone marrow-derived group 2 innate lymphoid cells. (b) Bone marrow cells were stained for Sca-1, CD117 (c-Kit), CD25, and for lineage markers (TCRβ, TCRγδ, CD3ɛ, Gr-1, CD11b, TER-119, B220, NK1.1, CD5, CD11c) and lineage-negative Sca-1+c-Kit-CD25+ group 2 innate lymphoid cells were enriched by flow cytometric cell sorting. (c) Purity and (d) expression of CD127, T1/ST2, ICOS, KLRG1, CD90.2 and Gata3 of sorted bone marrow-derived ILC2 was analyzed by flow cytometry. Cells were then cultured in the presence of IL-2, IL-7, IL-25, IL-33 (all at 50 ng/ml), and TSLP (20 ng/ml) for 15 days. Cells were given fresh medium and cytokines every two days and split into multiple wells when necessary to avoid excessive crowding, yielding an increase in ILC2 numbers of approximately 500-fold. The resulting cells were then seeded in fresh medium and allowed to rest in IL-2 and IL-7 (both at 10 ng/ml) for two days. Medium was removed from wells and rested cells were stimulated as necessary for up to five days.

Supplementary Figure 4 Type I interferon–mediated inhibition of the proliferation and cytokine production of ILC2 cells occurs independently of iNOS and STAT6.

Bone marrow cells from WT, inducible nitric oxide synthase (iNos)-deficient or Stat6-deficient mice were stimulated for five days in medium as a control or with IL‐7 and IL-33 (both at 10 ng/ml). IFN-β (25 or 500 U/ml) was added to some wells, as indicated. Proliferation of cells was measured by alamarBlue assay and cytokine production was assessed by ELISA of cell culture supernatants. The bars represent the mean ± standard deviation of triplicates of each cohort. Data are representative of three independent experiments. RFU, relative fluorescence units; n.d., not detectable; ns, not statistically significant (p > 0.05). The asterisks indicate statistically significant differences (*, p ≤ 0.05; **, p ≤ 0.01; ***, p ≤ 0.001).

Supplementary Figure 5 Type I interferon restrains mouse ILC2 cells.

(a,b) Sorted and expanded bone marrow-derived ILC2 from WT mice were stimulated for five days with IL‐7 and IL-33 (both at 10 ng/ml) only (black bars) or with IL‐7 and IL-33 (both at 10 ng/ml) in combination with IFN-β (500 U/ml; red bars). (a) IFN-β was added at different time points (0h, 24h, 48h and 72h) and left in the culture until supernatants were taken. (b) IFN-β was added at the beginning of the culture (0h) and at different time points (not removed, 24h, 48h and 72h) cells were washed and re-seeded with IL‐7 and IL-33 (both at 10 ng/ml) only until supernatants were taken. Five days after culture proliferation of cells was measured by alamarBlue assay and cytokine production was determined by ELISA. The percentages of reduction due to the addition of IFN-β are shown in brackets above the red bars. Data are representative of two independent experiments. (c) Sorted and expanded bone marrow-derived ILC2 from WT mice were stimulated for three days with medium only, or with IL‐7 and IL-33 (both at 10 ng/ml). IFN-β (25 or 500 U/ml) was added to some wells as indicated. GATA3 expression was determined by intracellular flow cytometry staining and histograms and the ratio of the GATA3 mean fluorescence intensities (MFI) to the MFI of the internal isotype control stainings are shown. Data are representative of three independent experiments. (d) Sorted and expanded bone marrow-derived ILC2 from WT mice were stimulated for five days with IL‐7 and IL-33 (both at 10 ng/ml) only (black bars) or with IL‐7 and IL-33 (both at 10 ng/ml) in combination with IFN-β (500 U/ml; red bars). IFN-β was added at the beginning of the culture (0h) and at different time points (24h, 48h and 72h) cells were washed and re-seeded with IL‐7 and IL-33 (both at 10 ng/ml) only until supernatants were taken. Five days after culture GATA3 expression was determined by intracellular flow cytometry staining and the ratio of the GATA3 MFI to the MFI of the internal isotype control stainings are shown. Data are representative of two independent experiments. The bars represent the mean ± standard deviation of triplicates of each cohort. The asterisks indicate statistically significant differences (*, p ≤ 0.05; **, p ≤ 0.01; ***, p ≤ 0.001). RFU, relative fluorescence units.

Supplementary Figure 6 Type I interferon restrains IL-33-mediated inflammation.

(a) Schematic model of cytokine administration protocol. WT mice (five age- and sex-matched animals per group) were challenged intranasally with PBS as control, IFNβ, IL-33, or IL-33 + IFN-β for three consecutive days. 24 hours after the last treatment pulmonary expression of (b) Il6, (c) Ccl17, (d) Ccl22, (e) Chi3l3, (f) Fn1 and (g) Ccl24 was determined by qRT-PCR. Data are representative of two independent experiments. (h,i) WT mice (five age- and sex-matched animals per group) were challenged intranasally with phosphate buffered saline (PBS) as a control, IFN-β, IL-33, or IL-33 + IFN-β for three consecutive days. 24 hours after the last treatment pulmonary cytokine expression and cellular composition of lungs were analyzed. (h) Expression of cell surface CD11c of pulmonary eosinophils was determined by flow cytometry. After setting singlet gate, viable eosinophils were further defined as SiglecF+CD11c-. The frequencies of eosinophils are highlighted in the red gates. (i) The mean fluorescence intensities (MFI) of cell surface CD11c expression by eosinophils are shown. The bars represent the mean ± standard deviation of each cohort. Data are representative of two independent experiments. The asterisks indicate statistically significant differences (*, p ≤ 0.05; **, p ≤ 0.01; ***, p ≤ 0.001).

Supplementary Figure 7 IFN-γ-mediated inhibition of the proliferation and cytokine production of ILC2 cells occurs independently of STAT2, IRF9, IFNAR, STAT4, T-bet, iNOS and STAT6.

Bone marrow cells from (a) WT and Stat2-/-, (b) WT and Irf9-/-, (c) WT and Ifnar1-/-, (d) WT and Stat4-mutant, (e) WT and Tbx21-/-, (f) WT, iNos-/- and Stat6-/- mice were stimulated for five days in medium as a control or with IL‐7 and IL-33 (both at 10 ng/ml). IFN-γ (5 or 20 ng/ml) was added to some wells, as indicated. Proliferation of cells was measured by alamarBlue assay, cytokine production was determined by ELISA of cell culture supernatants. The bars represent the mean ± standard deviation of triplicates of each cohort. Data are representative of three independent experiments. RFU, relative fluorescence units; n.d., not detectable; ns, not statistically significant (p > 0.05). The asterisks indicate statistically significant differences (*, p ≤ 0.05; **, p ≤ 0.01; ***, p ≤ 0.001).

Supplementary Figure 8 Type II interferon restrains IL-33-mediated inflammation.

Wild type (WT) C57BL/6 mice (five age- and sex-matched animals per group) were challenged intranasally with PBS as control, IFN-γ, IL-33, or IL-33 + IFN-γ for three consecutive days. 24 hours after the last treatment pulmonary expression of (a) Il6, (b) Ccl17, (c) Ccl22, (d) Chi3l3, (e) Fn1 and (f) Ccl24 was determined by qRT-PCR. The bars represent the mean ± standard deviation of each cohort. Data are representative of two independent experiments. The asterisks indicate statistically significant differences (*, p ≤ 0.05; **, p ≤ 0.01; ***, p ≤ 0.001).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1 and 2 (PDF 1799 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Duerr, C., McCarthy, C., Mindt, B. et al. Type I interferon restricts type 2 immunopathology through the regulation of group 2 innate lymphoid cells. Nat Immunol 17, 65–75 (2016). https://doi.org/10.1038/ni.3308

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3308

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