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:

Interferon-λ enhances adaptive mucosal immunity by boosting release of thymic stromal lymphopoietin

Abstract

Interferon-λ (IFN-λ) acts on mucosal epithelial cells and thereby confers direct antiviral protection. In contrast, the role of IFN-λ in adaptive immunity is far less clear. Here, we report that mice deficient in IFN-λ signaling exhibited impaired CD8+ T cell and antibody responses after infection with a live-attenuated influenza virus. Virus-induced release of IFN-λ triggered the synthesis of thymic stromal lymphopoietin (TSLP) by M cells in the upper airways that, in turn, stimulated migratory dendritic cells and boosted antigen-dependent germinal center reactions in draining lymph nodes. The IFN-λ–TSLP axis also boosted production of the immunoglobulins IgG1 and IgA after intranasal immunization with influenza virus subunit vaccines and improved survival of mice after challenge with virulent influenza viruses. IFN-λ did not influence the efficacy of vaccines applied by subcutaneous or intraperitoneal routes, indicating that IFN-λ plays a vital role in potentiating adaptive immune responses that initiate at mucosal surfaces.

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

Fig. 1: Defective immune response in Ifnlr1–/– mice after infection with live-attenuated influenza virus is not rescued by hematopoietic cells from wild type mice.
Fig. 2: IFN-λ enhances IgG1 and IgA production after intranasal application of influenza subunit vaccines.
Fig. 3: The immune-enhancing effect of IFN-λ depends on TSLP.
Fig. 4: Upper airway M cells produce TSLP after IFN-λ stimulation.
Fig. 5: IFN-λ promotes TSLP-dependent germinal center reactions.
Fig. 6: IFN-λ-triggered TSLP acts on CD103+ migratory DCs.
Fig. 7: Mucosal immunization in the presence of IFN-λ results in enhanced protection from influenza virus-induced disease.
Fig. 8: IFN-λ-adjuvanted M2e vaccine reduces influenza virus transmission.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon request.

References

  1. Crotta, S. et al. Type I and type III interferons drive redundant amplification loops to induce a transcriptional signature in influenza-infected airway epithelia. PLoS Pathog. 9, e1003773 (2013).

    Article  Google Scholar 

  2. Wack, A., Terczynska-Dyla, E. & Hartmann, R. Guarding the frontiers: the biology of type III interferons. Nat. Immunol. 16, 802–809 (2015).

    Article  CAS  Google Scholar 

  3. Lazear, H. M., Nice, T. J. & Diamond, M. S. Interferon-lambda: immune functions at barrier surfaces and beyond. Immunity 43, 15–28 (2015).

    Article  CAS  Google Scholar 

  4. Sommereyns, C., Paul, S., Staeheli, P. & Michiels, T. IFN-lambda (IFN-lambda) is expressed in a tissue-dependent fashion and primarily acts on epithelial cells in vivo. PLoS Pathog. 4, e1000017 (2008).

    Article  Google Scholar 

  5. Blazek, K. et al. IFN-lambda resolves inflammation via suppression of neutrophil infiltration and IL-1beta production. J. Exp. Med. 212, 845–853 (2015).

    Article  CAS  Google Scholar 

  6. Broggi, A., Tan, Y., Granucci, F. & Zanoni, I. IFN-lambda suppresses intestinal inflammation by non-translational regulation of neutrophil function. Nat. Immunol. 18, 1084–1093 (2017).

    Article  CAS  Google Scholar 

  7. de Groen, R. A., Groothuismink, Z. M., Liu, B. S. & Boonstra, A. IFN-lambda is able to augment TLR-mediated activation and subsequent function of primary human B cells. J. Leukoc. Biol. 98, 623–630 (2015).

    Article  Google Scholar 

  8. Espinoza, V. et al. Type III interferon is a critical regulator of innate antifungal immunity. Sci. Immunol. 2, eaan5357 (2017).

    Article  Google Scholar 

  9. Galani, I. E. et al. Interferon-lambda mediates non-redundant front-line antiviral protection against influenza virus infection without compromising host fitness. Immunity 46, 875–890 e876 (2017).

    Article  CAS  Google Scholar 

  10. Mordstein, M. et al. Lambda interferon renders epithelial cells of the respiratory and gastrointestinal tracts resistant to viral infections. J. Virol. 84, 5670–5677 (2010).

    Article  CAS  Google Scholar 

  11. Hernandez, P. P. et al. Interferon-lambda and interleukin 22 act synergistically for the induction of interferon-stimulated genes and control of rotavirus infection. Nat. Immunol. 16, 698–707 (2015).

    Article  CAS  Google Scholar 

  12. Mahlakoiv, T., Hernandez, P., Gronke, K., Diefenbach, A. & Staeheli, P. Leukocyte-derived IFN-alpha/beta and epithelial IFN-lambda constitute a compartmentalized mucosal defense system that restricts enteric virus infections. PLoS Pathog. 11, e1004782 (2015).

    Article  Google Scholar 

  13. Pott, J. et al. IFN-lambda determines the intestinal epithelial antiviral host defense. Proc. Natl Acad. Sci. USA 108, 7944–7949 (2011).

    Article  CAS  Google Scholar 

  14. Egli, A. et al. IL-28B is a key regulator of B- and T-cell vaccine responses against influenza. PLoS Pathog. 10, e1004556 (2014).

    Article  Google Scholar 

  15. Morrow, M. P. et al. Comparative ability of IL-12 and IL-28B to regulate Treg populations and enhance adaptive cellular immunity. Blood 113, 5868–5877 (2009).

    Article  CAS  Google Scholar 

  16. Zhou, Y. et al. Optimized DNA vaccine enhanced by adjuvant IL28B induces protective immune responses against herpes simplex virus type 2 in mice. Viral Immunol. 30, 601–614 (2017).

    Article  CAS  Google Scholar 

  17. Ferko, B. et al. Immunogenicity and protection efficacy of replication-deficient influenza A viruses with altered NS1 genes. J. Virol. 78, 13037–13045 (2004).

    Article  CAS  Google Scholar 

  18. Kochs, G. et al. Strong interferon-inducing capacity of a highly virulent variant of influenza A virus strain PR8 with deletions in the NS1 gene. J. Gen. Virol. 90, 2990–2994 (2009).

    Article  CAS  Google Scholar 

  19. Ank, N. et al. An important role for type III interferon (IFN-lambda/IL-28) in TLR-induced antiviral activity. J. Immunol. 180, 2474–2485 (2008).

    Article  CAS  Google Scholar 

  20. Dellgren, C., Gad, H. H., Hamming, O. J., Melchjorsen, J. & Hartmann, R. Human interferon-lambda3 is a potent member of the type III interferon family. Genes Immun. 10, 125–131 (2009).

    Article  CAS  Google Scholar 

  21. Eliasson, D. G. et al. CTA1-M2e-DD: a novel mucosal adjuvant targeted influenza vaccine. Vaccine 26, 1243–1252 (2008).

    Article  CAS  Google Scholar 

  22. Van Roey, G. A., Arias, M. A., Tregoning, J. S., Rowe, G. & Shattock, R. J. Thymic stromal lymphopoietin (TSLP) acts as a potent mucosal adjuvant for HIV-1 gp140 vaccination in mice. Eur. J. Immunol. 42, 353–363 (2012).

    Article  Google Scholar 

  23. Chappaz, S., Flueck, L., Farr, A. G., Rolink, A. G. & Finke, D. Increased TSLP availability restores T- and B-cell compartments in adult IL-7 deficient mice. Blood 110, 3862–3870 (2007).

    Article  CAS  Google Scholar 

  24. Ziegler, S. F. & Artis, D. Sensing the outside world: TSLP regulates barrier immunity. Nat. Immunol. 11, 289–293 (2010).

    Article  CAS  Google Scholar 

  25. Joo, S. et al. Critical role of TSLP-responsive mucosal dendritic cells in the induction of nasal antigen-specific IgA response. Mucosal Immunol. 10, 901–911 (2017).

    Article  CAS  Google Scholar 

  26. Soumelis, V. et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat. Immunol. 3, 673–680 (2002).

    Article  CAS  Google Scholar 

  27. Kim, D. Y. et al. The airway antigen sampling system: respiratory M cells as an alternative gateway for inhaled antigens. J. Immunol. 186, 4253–4262 (2011).

    Article  CAS  Google Scholar 

  28. Vinuesa, C. G., Linterman, M. A., Yu, D. & MacLennan, I. C. M. Follicular helper T cells. Annu. Rev. Immunol. 34, 335–368 (2016).

    Article  CAS  Google Scholar 

  29. Fazilleau, N., Mark, L., McHeyzer-Williams, L. J. & McHeyzer-Williams, M. G. Follicular helper T cells: lineage and location. Immunity 30, 324–335 (2009).

    Article  CAS  Google Scholar 

  30. Grajales-Reyes, G. E. et al. Batf3 maintains autoactivation of Irf8 for commitment of a CD8alpha(+) conventional DC clonogenic progenitor. Nat. Immunol. 16, 708–717 (2015).

    Article  CAS  Google Scholar 

  31. Takaki, H. et al. Toll-like receptor 3 in nasal CD103(+) dendritic cells is involved in immunoglobulin A production. Mucosal Immunol. 11, 82–96 (2018).

    Article  CAS  Google Scholar 

  32. Van den Hoecke, S. et al. Hierarchical and redundant roles of activating FcgammaRs in protection against influenza disease by M2e-Specific IgG1 and IgG2a antibodies. J. Virol. 91, e02500–e02516 (2017).

    PubMed  PubMed Central  Google Scholar 

  33. Grimm, D. et al. Replication fitness determines high virulence of influenza A virus in mice carrying functional Mx1 resistance gene. Proc. Natl Acad. Sci. USA 104, 6806–6811 (2007).

    Article  CAS  Google Scholar 

  34. Klinkhammer, J. et al. IFN-lambda prevents influenza virus spread from the upper airways to the lungs and limits virus transmission. Elife 7, e33354 (2018).

    Article  Google Scholar 

  35. Kolpe, A., Schepens, B., Ye, L., Staeheli, P. & Saelens, X. Passively transferred M2e-specific monoclonal antibody reduces influenza A virus transmission in mice. Antiviral Res. 158, 244–254 (2018).

    Article  CAS  Google Scholar 

  36. Mordstein, M. et al. Interferon-lambda contributes to innate immunity of mice against influenza A virus but not against hepatotropic viruses. PLoS Pathog. 4, e1000151 (2008).

    Article  Google Scholar 

  37. Lee, H. C. et al. Thymic stromal lymphopoietin is induced by respiratory syncytial virus-infected airway epithelial cells and promotes a type 2 response to infection. J. Allergy Clin. Immunol. 130, 1187–1196.e1185 (2012).

    Article  CAS  Google Scholar 

  38. Ganti, K. P., Mukherji, A., Surjit, M., Li, M. & Chambon, P. Similarities and differences in the transcriptional control of expression of the mouse TSLP gene in skin epidermis and intestinal epithelium. Proc. Natl Acad. Sci. USA 114, E951–E960 (2017).

    Article  CAS  Google Scholar 

  39. Kelly, A. et al. Immune cell profiling of IFN-lambda response shows pDCs express highest level of IFN-lambdaR1 and are directly responsive via the JAK-STAT pathway. J. Interferon Cytokine Res. 36, 671–680 (2016).

    Article  CAS  Google Scholar 

  40. West, E. E. et al. A TSLP-complement axis mediates neutrophil killing of methicillin-resistant Staphylococcus aureus. Sci. Immunol. 1, eaaf8471 (2016).

    Article  Google Scholar 

Download references

Acknowledgements

We thank A. Ohnemus for technical support, and H. Pircher, G. Gasteiger and O. Haller for helpful discussions and comments on the manuscript. Funding for this work was provided by the European Union’s Seventh Framework Program grant agreement 607690 (to P.S. and N.L.), the Deutsche Forschungsgemeinschaft grant agreement STA 338/15-1 (to P.S.) and TA 436/4-1 (to Y.T.), the Else Kröner-Fresenius Stiftung grant agreement 2017_EKES.34 (to Y.T.) and the Danish Council for Independent Research, Medical Research grant agreement 11‐107588 (to R.H.).

Author information

Authors and Affiliations

Authors

Contributions

L.Y. designed and performed most of the experiments, analyzed and interpreted the data. D.S. designed and performed experiments, analyzed and interpreted the data. J.B. performed experiments and analyzed samples. K.E. and D.S. generated bone marrow chimeric mice. V.B. and H.H.G. provided essential materials. Y.T., R.H. and N.L. analyzed and interpreted data, and gave advice. P.S. conceived the project, acquired funding for the study, designed experiments and interpreted the data. P.S., L.Y. and D.S. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Peter Staeheli.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 IFN-λ exhibits no immunostimulatory effect when antigen is applied by the intraperitoneal or subcutaneous route.

a-b, Titers of HA-specific IgG subclasses on day 10 after the first and second booster immunization in sera of Mx1-WT mice immunized by the intraperitoneal (a) or subcutaneous (b) route with Influsplit Tetra® in the presence or absence of IFN-λ2. Data are representative of two independent experiments with n=6 (a) and n=7 (b) animals per group. Error bars represent SEM centered on the mean. Each symbol represents an individual animal. c-d, Titers of M2e-specific IgG subclasses on day 10 post booster immunization in sera of Mx1-WT mice immunized by either the intraperitoneal (c) or subcutaneous (d) route with M2e vaccine in the presence or absence of IFN-λ2. Data are representative of two independent experiments with n=6 animals per group. Error bars represent SEM centered on the mean. Each symbol represents an individual animal.

Supplementary Figure 2 IFN-λ-triggered enhanced antibody synthesis depends on TSLP produced by upper airway M cells.

a, upper panels, Representative flow cytometry histograms of TSLP staining in cell subsets from snouts of Mx1-WT and Mx1-Ifnlr1–/– mice infected with hvPR8-ΔNS1 for 24 h. a, lower panels, MFI of TSLP signals in Mx1-WT (n=6 per group) and Mx1-Ifnlr1–/– mice (n=6 in mock group, n=5 in infected group) infected with hvPR8-ΔNS1 for 24 h. Data are pooled from two independent experiments. Error bars represent SEM centered on the mean. **p=0.01, *p=0.0166 by unpaired two-tailed Student’s . b, upper panels, Representative flow cytometry histograms of TSLP staining in cell subsets from snouts of Mx1-WT and Mx1-Ifnlr1–/– mice treated with IFN-λ2 for 24 h. b, lower panels, MFI of TSLP signals in Mx1-WT (n=6 in mock group and n=9 in infected group) and Mx1-Ifnlr1–/– mice (n=7 per group) treated with IFN-λ2 for 24 h. Data are pooled from two independent experiments. Error bars represent SEM centered on the mean. ****p<0.0001 by unpaired two-tailed Student’s t test. c, Levels of Tslp and Mx1 mRNA in purified snout cell subsets from IFN-λ2-treated Mx1-WT mice. Cells originating from 12 animals per group were pooled prior to cell sorting. Data are pooled from three independent experiments. Error bars represent SEM centered on the mean. *p=0.0252 by unpaired two-tailed Student’s t test.

Supplementary Figure 3 Gating strategy used to isolate M cells from the upper airways of mice.

a, Snout cells were gated based on forward and side scatters, then single cells, live cells, CD45+ cells (CD45+ EpCAM-), epithelial cells (CD45- EpCAM+), M+ cells (CD45- EpCAM+ NKM16-2-4+) and M- cells (CD45- EpCAM+ NKM16-2-4-) were gated. b, Strategy for sorting CD45+ cells (CD45+ EpCAM-, 99% purity), M+ cells (CD45- EpCAM+ NKM16-2-4+, 93.9% purity) and M- cells (CD45- EpCAM+ NKM16-2-4-, 98.9% purity) from Mx1-WT mice treated with IFN-λ2 for 4 h.

Supplementary Figure 4 M cells in mouse airway epithelial cells at the air–liquid interface of cultures do not substantially upregulate TSLP synthesis upon stimulation with IFN-λ.

a-b, In vitro differentiated airway epithelial cells from Mx1-WT mice were treated with or without IFN-λ2 for 24 h before expression of TSLP in M+ (EpCAM+ NKM16-2-4+) (a) and M- (EpCAM+ NKM16-2-4-) cells (b) was analyzed by flow cytometry. Each symbol represents an individual replicate. a-b, n=7 and n=9 independent cell culture wells for mock and IFN-λ2-treated groups, respectively. Data are pooled from two individual experiments and shown as mean ± SEM. *p=0.0149 (a) and p=0.0146 (b) by unpaired two-tailed Student’s t test.

Supplementary Figure 5 IFN-λ increases the frequency of Tfh cells and GC-B cells in the spleen of immunized mice in a TSLP-dependent manner.

a-c, WT (n=3 for M2e group and n=4 for M2e+IFN-λ group) and Tslpr–/– (n=4 per group) mice intranasally immunized with M2e in the presence or absence of IFN-λ2 were sacrificed on day 5 post boosting, and the frequencies of CXCR5+ PD1+ Tfh cells among CD19- CD4+ CD44+ cells (a), GL7+ Fas+ GC-B cells among CD4- B220+ cells (b) and IgG1+ Fas+ GC-B cells among CD4- B220+ cells (c) in the spleen were determined. a-c, left panels, Representative flow cytometry histograms. a-c, right panels, data are from one of two representative experiments. Each symbol represents an individual animal. Shown are mean ± SEM. ****P<0.0001 (a, left), ****P<0.0001 (a, right), **p=0.0012 (b, left), **p=0.0011 (b, right), ***p=0.0001 (c, left), ***p=0.0006 (c, right) by unpaired two-tailed Student’s t-test.

Supplementary Figure 6 IFN-λ does not induce GC reactions in the absence of antigen.

a-c, WT and Tslpr–/– mice were mock-immunized in the presence or absence of IFN-λ2. Animals were sacrificed on day 5 post boosting, and the frequencies of CXCR5+ PD1+ Tfh cells among CD19- CD4+ CD44+ cells (a), GL7+ Fas+ GC-B cells among CD4- B220+ cells (b) and IgG1+ Fas+ GC-B cells (c) among CD4- B220+ cells in the spleen and draining LN were determined. a, n=5 mice per group. b-c, left panel, n=5 mice per group. b-c, right panel, n=4 in mock group, n=5 in IFN-λ group. Each symbol represents an individual animal. Error bars represent SEM centered on the mean.

Supplementary Figure 7 Mucosal immunization in the presence of IFN-λ results in enhanced suppression of the replication of influenza virus in lungs.

Mx1-WT mice were intranasally immunized with M2e vaccine in the presence (n=7) or absence (n=6) of IFN-λ2. Ten days after booster immunization, the animals were challenged with hvPR8. Viral titers were measured by plaque assay at day 2 post-infection. Mock-immunized mice (PBS) served as additional controls (n=6). Each symbol represents an individual animal. Data are shown as mean ± SEM. ****p<0.0001 by one-way ANOVA with Tukey’s multiple-comparison test.

Supplementary Figure 8 TSLP-adjuvanted M2e vaccine reduces the transmission of influenza virus from infected mice to naive cage mates.

BALB/c mice were intranasally immunized with M2e vaccine in the presence (n=6) or absence (n=6) of TSLP. Mock-immunized animals (PBS) served as additional controls (n=3). Eight weeks after booster immunization, the animals were infected with influenza virus strain A/Udorn. Twenty-four hours later, each infected BALB/c mouse was co-housed with one naive DBA/2J mouse for 4 days. Virus transmission to contact mice was assessed by measuring infectious virus in the snouts of immunized BALB/c (Index) and exposed DBA/2J mice (Contact) on day 4 post cohousing. Each symbol represents an individual animal. Small horizontal lines indicated the mean. The dotted line indicates the detection limit of the assay.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ye, L., Schnepf, D., Becker, J. et al. Interferon-λ enhances adaptive mucosal immunity by boosting release of thymic stromal lymphopoietin. Nat Immunol 20, 593–601 (2019). https://doi.org/10.1038/s41590-019-0345-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0345-x

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