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:

IFNγ-induction of TH1-like regulatory T cells controls antiviral responses

Abstract

Regulatory T (Treg) cells are an immunosuppressive population that are required to maintain peripheral tolerance and prevent tissue damage from immunopathology, via anti-inflammatory cytokines, inhibitor receptors and metabolic disruption. Here we show that Treg cells acquire an effector-like state, yet remain stable and functional, when exposed to interferon gamma (IFNγ) during infection with lymphocytic choriomeningitis and influenza A virus. Treg cell-restricted deletion of the IFNγ receptor (encoded by Ifngr1), but not the interleukin 12 (IL12) receptor (encoded by Il12rb2), prevented TH1-like polarization (decreased expression of T-bet, CXC motif chemokine receptor 3 and IFNγ) and promoted TH2-like polarization (increased expression of GATA-3, CCR4 and IL4). TH1-like Treg cells limited CD8+ T cell effector function, proliferation and memory formation during acute and chronic infection. These findings provide fundamental insights into how Treg cells sense inflammatory cues from the environment (such as IFNγ) during viral infection to provide guidance to the effector immune response. This regulatory circuit prevents prolonged immunoinflammatory responses and shapes the quality and quantity of the memory T cell response.

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: Treg cells acquire a TH1-like program during acute and chronic infection.
Fig. 2: Transcriptomic analysis reveals IFNγ-induced TH1 profiles in Treg cells during chronic infection.
Fig. 3: Ifngr1-deficient Treg cells program to a TH2 phenotype during viral infection.
Fig. 4: IL12 does not induce TH1-like nor suppress TH2-like programming during chronic infection.
Fig. 5: TH1-like Treg cells limit T cell effector function during chronic infection.
Fig. 6: IFNγ from CD8+ T cells but not Treg cells contributes to TH1 polarization of Treg cells during chronic infection.
Fig. 7: TH1-like Treg cells limit CD8+ T memory formation and response to vaccination during acute and chronic infection.

Similar content being viewed by others

Data availability

The RNA sequencing data that support the findings of this study have been deposited in the GEO under the accession code GSE223210. All other data are present in the article and Supplementary files or from the corresponding author upon reasonable request. Source data files are present. Source data are provided with this paper.

References

  1. Veiga-Parga, T., Sehrawat, S. & Rouse, B. T. Role of regulatory T cells during virus infection. Immunol. Rev. 255, 182–196 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Punkosdy, G. A. et al. Regulatory T-cell expansion during chronic viral infection is dependent on endogenous retroviral superantigens. Proc. Natl Acad. Sci. USA 108, 3677–3682 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Cabrera, R. et al. An immunomodulatory role for CD4+CD25+ regulatory T lymphocytes in hepatitis C virus infection. Hepatology 40, 1062–1071 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Betts, R. J., Ho, A. W. S. & Kemeny, D. M. Partial depletion of natural CD4+CD25+ regulatory T cells with anti-CD25 antibody does not alter the course of acute influenza A virus infection. PLoS ONE 6, e27849 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Penaloza-MacMaster, P. et al. Interplay between regulatory T cells and PD-1 in modulating T cell exhaustion and viral control during chronic LCMV infection. J. Exp. Med. 211, 1905–1918 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Saravia, J., Chapman, N. M. & Chi, H. Helper T cell differentiation. Cell Mol. Immunol. 16, 634–643 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Zhu, J. et al. The transcription factor T-bet is induced by multiple pathways and prevents an endogenous Th2 cell program during Th1 cell responses. Immunity 37, 660–673 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lazarevic, V. et al. T-bet represses T(H)17 differentiation by preventing Runx1-mediated activation of the gene encoding RORγt. Nat. Immunol. 12, 96–104 (2011).

    Article  CAS  PubMed  Google Scholar 

  9. Verma, N. D. et al. Interleukin-12 (IL-12p70) promotes induction of highly potent Th1-like CD4+CD25+ T regulatory cells that inhibit allograft rejection in unmodified recipients. Front. Immunol. 5, 190 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Cousens, L. P., Orange, J. S., Su, H. C. & Biron, C. A. Interferon-alpha/beta inhibition of interleukin 12 and interferon-gamma production in vitro and endogenously during viral infection. Proc. Natl Acad. Sci. USA 94, 634–639 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Byrnes, A. A. et al. Type I interferons and IL-12: convergence and cross-regulation among mediators of cellular immunity. Eur. J. Immunol. 31, 2026–2034 (2001).

    Article  CAS  PubMed  Google Scholar 

  12. Levine, A. G. et al. Stability and function of regulatory T cells expressing the transcription factor T-bet. Nature 546, 421–425 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Overacre-Delgoffe, A. E. et al. Interferon-γ drives treg fragility to promote anti-tumor immunity. Cell 169, 1130–1141 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Rubtsov, Y. P. et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546–558 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Smeltz, R. B., Chen, J., Ehrhardt, R. & Shevach, E. M. Role of IFN-gamma in Th1 differentiation: IFN-gamma regulates IL-18R alpha expression by preventing the negative effects of IL-4 and by inducing/maintaining IL-12 receptor beta 2 expression. J. Immunol. 168, 6165–6172 (2002).

    Article  CAS  PubMed  Google Scholar 

  16. Naka, T. et al. SOCS-1/SSI-1-deficient NKT cells participate in severe hepatitis through dysregulated cross-talk inhibition of IFN-gamma and IL-4 signaling in vivo. Immunity 14, 535–545 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Lee, S. H. et al. Identifying the initiating events of anti-Listeria responses using mice with conditional loss of IFN-γ receptor subunit 1 (IFNGR1). J. Immunol. 191, 4223–4234 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Yang, X., Bam, M., Becker, W., Nagarkatti, P. S. & Nagarkatti, M. Long noncoding RNA AW112010 promotes the differentiation of inflammatory T cells by suppressing IL-10 expression through histone demethylation. J. Immunol. 205, 987–993 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Marshall, H. D. et al. Differential expression of Ly6C and T-bet distinguish effector and memory Th1 CD4+ cell properties during viral infection. Immunity 35, 633–646 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Heninger, A.-K. et al. IL-7 abrogates suppressive activity of human CD4+CD25+FOXP3+ regulatory T cells and allows expansion of alloreactive and autoreactive T cells. J. Immunol. 189, 5649–5658 (2012).

    Article  CAS  PubMed  Google Scholar 

  21. Wei, G. et al. Genome-wide analyses of transcription factor GATA3-mediated gene regulation in distinct T cell types. Immunity 35, 299–311 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Schiering, C. et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature 513, 564–568 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Stubbington, M. J. et al. An atlas of mouse CD4+ T cell transcriptomes. Biol. Direct 10, 14 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Briggs, S. F. & Reijo Pera, R. A. X chromosome inactivation: recent advances and a look forward. Curr. Opin. Genet. Dev. 28, 78–82 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Rubtsov, Y. P. et al. Stability of the regulatory T cell lineage in vivo. Science 329, 1667–1671 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Baazim, H. et al. CD8+ T cells induce cachexia during chronic viral infection. Nat. Immunol. 20, 701–710 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zinkernagel, R. M. et al. T cell-mediated hepatitis in mice infected with lymphocytic choriomeningitis virus. Liver cell destruction by H-2 class I-restricted virus-specific cytotoxic T cells as a physiological correlate of the 51Cr-release assay? J. Exp. Med. 164, 1075–1092 (1986).

    Article  CAS  PubMed  Google Scholar 

  28. Zajac, A. J. et al. Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med. 188, 2205–2213 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wherry, E. J., Blattman, J. N., Murali-Krishna, K., van der Most, R. & Ahmed, R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J. Virol. 77, 4911–4927 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Liu, C. et al. Treg cells promote the SREBP1-dependent metabolic fitness of tumor-promoting macrophages via repression of CD8+ T cell-derived interferon-γ. Immunity 51, 381–397 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ferreira, C. et al. Type 1 Treg cells promote the generation of CD8+ tissue-resident memory T cells. Nat. Immunol. 21, 766–776 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Pace, L. et al. Regulatory T cells increase the avidity of primary CD8+ T cell responses and promote memory. Science 338, 532–536 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Bhattacharyya, M. & Penaloza-MacMaster, P. T regulatory cells are critical for the maintenance, anamnestic expansion and protection elicited by vaccine-induced CD8 T cells. Immunology 151, 340–348 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kaech, S. M., Hemby, S., Kersh, E. & Ahmed, R. Molecular and functional profiling of memory CD8 T cell differentiation. Cell 111, 837–851 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Franckaert, D. et al. Promiscuous Foxp3-cre activity reveals a differential requirement for CD28 in Foxp3+ and Foxp3 T cells. Immunol. Cell Biol. 93, 417–423 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Bittner-Eddy, P. D., Fischer, L. A. & Costalonga, M. Cre-loxP reporter mouse reveals stochastic activity of the Foxp3 promoter. Front. Immunol. 10, 2228 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Martin, M. D. & Badovinac, V. P. Defining memory CD8 T cell. Front. Immunol. 9, 2692 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Littringer, K. et al. Common features of regulatory T cell specialization during Th1 responses. Front. Immunol. 9, 1344 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Lynch, E. A., Heijens, C. A. W., Horst, N. F., Center, D. M. & Cruikshank, W. W. Cutting edge: IL-16/CD4 preferentially induces Th1 cell migration: requirement of CCR5. J. Immunol. 171, 4965–4968 (2003).

    Article  CAS  PubMed  Google Scholar 

  40. Cipolla, E. M. et al. Heterotypic influenza infections mitigate susceptibility to secondary bacterial infection. J. Immunol. 209, 760–771 (2022).

    Article  CAS  PubMed  Google Scholar 

  41. Koch, M. A. et al. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat. Immunol. 10, 595–602 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. McFadden, C. et al. Preferential migration of T regulatory cells induced by IL-16. J. Immunol. 179, 6439–6445 (2007).

    Article  CAS  PubMed  Google Scholar 

  43. Oxenius, A., Karrer, U., Zinkernagel, R. M. & Hengartner, H. IL-12 is not required for induction of type 1 cytokine responses in viral infections. J. Immunol. 162, 965–973 (1999).

    Article  CAS  PubMed  Google Scholar 

  44. Ashour, D. et al. IL-12 from endogenous cDC1, and not vaccine DC, is required for Th1 induction. JCI Insight 5, e135143 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Cao, X. et al. Interleukin 12 stimulates IFN-gamma-mediated inhibition of tumor-induced regulatory T-cell proliferation and enhances tumor clearance. Cancer Res. 69, 8700–8709 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Solouki, S. et al. TCR signal strength and antigen affinity regulate CD8+ memory T cells. J. Immunol. 205, 1217–1227 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. Iborra, S. et al. Optimal generation of tissue-resident but not circulating memory T cells during viral infection requires crosspriming by DNGR-1+ dendritic cells. Immunity 45, 847–860 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Slütter, B., Pewe, L. L., Kaech, S. M. & Harty, J. T. Lung airway-surveilling CXCR3(hi) memory CD8(+) T cells are critical for protection against influenza A virus. Immunity 39, 939–948 (2013).

    Article  PubMed  Google Scholar 

  49. Ndure, J. & Flanagan, K. L. Targeting regulatory T cells to improve vaccine immunogenicity in early life. Front. Microbiol. 5, 477 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Safar, H. A., Mustafa, A. S., Amoudy, H. A. & El-Hashim, A. The effect of adjuvants and delivery systems on Th1, Th2, Th17 and Treg cytokine responses in mice immunized with Mycobacterium tuberculosis-specific proteins. PLoS ONE 15, e0228381 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Welsh, R. M. & Seedhom, M. O. Lymphocytic choriomeningitis virus (LCMV): propagation, quantitation, and storage. Curr. Protoc. Microbiol. 15, 15A.1 (2008).

    Google Scholar 

  52. Robinson, K. M. et al. Novel protective mechanism for interleukin-33 at the mucosal barrier during influenza-associated bacterial superinfection. Mucosal Immunol. 11, 199–208 (2018).

    Article  CAS  PubMed  Google Scholar 

  53. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  54. Shen, F. W. et al. Cloning of Ly-5 cDNA. Proc. Natl Acad. Sci. USA 82, 7360–7363 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Mombaerts, P. et al. RAG-1-deficient mice have no mature B and T lymphocytes. Cell 68, 869–877 (1992).

    Article  CAS  PubMed  Google Scholar 

  56. Snell, G. D., Cherry, M., McKenzie, I. F. & Bailey, D. W. Ly-4, a new locus determining a lymphocyte cell-surface alloantigen in mice. Proc. Natl. Acad. Sci. USA 70, 1108–1111 (1973).

  57. Liu, P., Jenkins, N. A. & Copeland, N. G. A highly efficient recombineering-based method for generating conditional knockout mutations. Genome Res. 13, 476–484 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Liu, C. et al. Neuropilin-1 is a T cell memory checkpoint limiting long-term antitumor immunity. Nat. Immunol. 21, 1010–1021 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Roederer, M., Nozzi, J. L. & Nason, M. C. SPICE: exploration and analysis of post-cytometric complex multivariate datasets. Cytometry A 79, 167–174 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Turnis, M. E. et al. Interleukin-35 limits anti-tumor immunity. Immunity 44, 316–329 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Schreiber, R. D. Measurement of mouse and human interferon gamma. Curr. Protoc. Immunol. 6, 6.8 (2001).

    Google Scholar 

  62. Buettner, F. et al. Computational analysis of cell-to-cell heterogeneity in single-cell RNA-sequencing data reveals hidden subpopulations of cells. Nat. Biotechnol. 33, 155–160 (2015).

    Article  CAS  PubMed  Google Scholar 

  63. He, Z., Brazovskaja, A., Ebert, S., Camp, J. G. & Treutlein, B. CSS: cluster similarity spectrum integration of single-cell genomics data. Genome Biol. 21, 224 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Korotkevich, G. et al. Fast gene set enrichment analysis. Preprint at bioRxiv https://doi.org/10.1101/060012 (2019).

  65. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation 2, 100141 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank everyone in the Vignali Lab (Vignali-lab.com; @Vignali_Lab) for all their constructive comments and advice. We thank R. Ahmed (Emory University) for providing parental LCMV viral stocks and protocols, S. Kaech (Yale University) for providing LM-GP33 stocks and protocols, L. D’Cruz and A. Piccirillo (University of Pittsburgh) for providing LM-OVA stocks and protocols, S. Canna, E. Rapp, P. Tsoukas and H. Nieves-Rosado (University of Pittsburgh) for growing up LCMV viral stocks, C. Workman for intravenous injections, E. Brunazzi for maintenance of mouse colonies, R. Dadey for helping with flow cytometry staining on LCMV-infected samples, C. Liu for aiding in flow cytometry panel design, C. Cardello for conducting libraries for scRNA-seq, H. Yano for constructing SPICE plots, D. Normolle for calculating statistics for the weight loss curves, L. Andrews for weighing mice and conducting intravenous injections, L. Rigatti for scoring livers, the University of Pittsburgh Biospecimen Core for histology, the University of Pittsburgh Innovative Technologies Development Core for developing new mouse strains, the University of Pittsburgh Center for Research Computing (HTC cluster) for next-generation sequencing and the University of Pittsburgh Flow Core for FACS and flow cytometry. This work was supported by the National Institutes of Health (F32 CA247004-01 and T32 CA082084 to A.M.G.D.; P01 AI108545, R35 CA263850 and R01 CA203689 to D.A.A.V.; R01 DK130897, R21 CA259636, P30 DK120531 and P30 CA047904 to M.M.; R01 HL107380 to J.F.A.; and R01 CA206517 and R01 AI138504 to L.P.K.). This work benefited from a SPECIAL ORDER BD LSR FORTESSA (funded by NIH 1S10 OD011925-01) used in the UPSOM Unified Flow Core and the University of Pittsburgh Center for Research Computing (funded by NIH S10OD028483).

Author information

Authors and Affiliations

Authors

Contributions

D.A.A.V. conceived, directed and obtained funding for the project. A.M.G.D., C.J.W. and D.A.A.V. conceptualized, designed and analyzed the experiments and wrote the manuscript. A.M.G.D. performed the majority of the experiments. J.C. performed the analysis of the scRNA-seq dataset. A.L.S.W., K.M.V. and C.J.W. generated new mouse strains. J.N.L., G.P.P and J.C.K. helped with processing the tissues before flow cytometry and FACS. J.N.L. helped with LM-GP33 experiments and plaque assays. G.P.P. validated mouse strains, performed qPCRs and isolated splenic T cells. J.C.K. performed qPCRs and isolated splenic T cells. L.A.A. and L.P.K. helped with the LCMV and LM-GP33 model and L.A.A. conducted some Arm. experiments. E.M.S. and B.R.H. infected mice with IAV and helped in necroscopy. L.H. and M.M. performed the ALT and AST activity assay. J.F.A. and C.J.W. contributed to experimental design and interpretation of the data. J.F.A. scored infected lungs. All authors provided feedback and approved the manuscript.

Corresponding author

Correspondence to Dario A. A. Vignali.

Ethics declarations

Competing interests

D.A.A.V. is a cofounder and stockholder for Novasenta, Potenza, Tizona and Trishula, is a stockholder for Oncorus, Werewolf and Apeximmune, holds patents licensed and royalties in BMS and Novasenta, serves on a scientific advisory board for Tizona, Werewolf, F-Star, Bicara, Apeximmune and T7/Imreg Bio and is a consultant for BMS, Incyte, G1 Therapeutics, Inzen Therapeutics, Regeneron and Avidity Partners. All the other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: N. Bernard managed the editorial process and peer review in collaboration with the rest of the editorial team of this article.

Additional information

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

Extended data

Extended Data Fig. 1 IFNγ and T-bet expression in Treg cells during acute and chronic infection.

a, Gating strategy for all flow cytometric experiments. b-f, Foxp3Cre-YFP mice remained uninfected (n = 5) or were infected (n = 6) with Arm. or Cl. 13. b, c, Expression of Ifng (b) and Tbx21 (c) mRNA by purified splenic Treg cells were determined by qPCR after ex vivo stimulation with anti-CD3/anti-CD28 (Arm.) or PMA and Ionomycin (Cl. 13). c, Linear regression of Tbx21 and Ifng co-expression. d, Foxp3Cre-YFP mice (n = 14) were infected with Arm. and stimulated ex vivo with anti-CD3 and expression of T-bet and IFNγ was measured by flow cytometry. Linear regression of T-bet and IFNγ co-expression. e, f, Foxp3Cre-YFP mice remained uninfected or were infected with Arm. or Cl. 13. Expression of Ifng (e) (n = 15, 5 independent experiments, Uninfected; n = 12, Arm.; n = 17 Cl. 13) and Tbx21 (f) (n = 16, 5 independent experiments, Uninfected; n = 12, Arm.; n = 14, Cl. 13) mRNA by sorted splenic CD4+ Tconv cells and Treg cells, without stimulation, were determined by qPCR. b-f, Data are presented as mean values and represent biologically independent mice and (b-d and as indicated) or 3 (e, f) or 5 (as indicated) independent experiments. Statistical significance was determined by multiple unpaired Student’s t-test relative to Uninfected (b, c), or by simple linear regression (d) or by a two-way ANOVA with multiple comparisons and multiple unpaired t-tests relative to CD4+ Tconv cells (e, f) (P values indicated when significant); NS, not significant.

Source data

Extended Data Fig. 2 TH1 signatures in Treg cells and CD4+ Tconv cells during acute and chronic infection.

a, Diagram of Il12rb2.Thy1.1 L/LhNGR targeted mouse. b, c, Il12rb2.Thy1.1 L/LhNGR mice (n = 11) were infected with Arm. and splenic Thy1.1 and Thy1.1+ CD4+ Tconv. cells purified and expression of Il12rb2 by Thy1.1 and Thy1.1+ CD4+ Tconv. cells was determined by qPCR (b). Thy1.1 and Thy1.1+ CD4+ Tconv. cells were serum starved and treated with PBS or IL-12, and induction of pSTAT4 was measured by flow cytometry (c). d, e, Foxp3Cre-YFP mice (n = 16, 5 independent experiments, Uninfected; n = 12, Arm.; n = 14, Cl. 13) remained uninfected or were infected with Arm. or Cl. 13, expression of Il12rb2 mRNA (d) and Ifngr1 mRNA (e) by sorted splenic CD4+ Tconv cells and Treg cells, without stimulation, were measured by qPCR. f-i, Expression of IFNGR1 (f) (n = 7, 2 independent experiments, Uninfected; n = 9, 2 independent experiments, Arm.; n = 15, Cl. 13), T-bet (g) (n = 7, Uninfected; n = 11, Arm.; n = 8, Cl. 13), CXCR3 (h) (n = 11, Uninfected; n = 9, 2 independent experiments, Arm.; n = 15, Cl. 13), T-bet (n = 7, 2 independent experiments, Uninfected; n = 11, 2 independent experiments, Arm.; n = 16, Cl. 13), GATA-3 (n = 6, 2 independent experiments, Uninfected; n = 9, 2 independent experiments, Arm.; n = 17, Cl. 13), and RORγt (n = 3, 2 independent experiments, Uninfected; n = 11, 2 independent experiments, Arm.; n = 8, 2 independent experiments, Cl. 13), (i) by splenic CD4+ Tconv cells and Treg cells were determined flow cytometry. b-i, Data are presented as mean values and represent biologically independent mice and 2 (g, and as indicated) or 3 (b-f, h, i) or 5 (as indicated) independent experiments. Statistical significance was determined by an unpaired two-tailed Student’s t-test relative to Thy1.1 (b) or CD4+ Tconv cells (f-h), or by a two-way AVOVA with multiple comparisons (c), or by a two-way ANOVA with multiple comparisons (d-e) and multiple unpaired t-tests relative to CD4+ Tconv cells (d, e, i) (P values indicated when significant); NS, not significant.

Source data

Extended Data Fig. 3 Transcriptomic analysis of Ifngr1-deficient Treg cells verses control, during chronic infection.

a, Expression of the deleted region of Ifngr1, normalized to the intact region of Ifngr1, from uninfected Ifngr1L/LFoxp3Cre-YFP (n = 5) compared to Foxp3Cre-YFP mice (n = 6). Thy1.2 cells, CD8+ T cells, CD4+ Tconv cells and Treg cells were sorted, gDNA was isolated and qPCR was performed. b, Expression of IFNGR1 in Treg cells from uninfected Foxp3Cre-YFP (n = 7) and Ifngr1L/LFoxp3Cre-YFP (n = 5) mice was measured by flow cytometry. c, d, Foxp3Cre-YFP mice remained uninfected (n = 15, 5 independent experiments, control; n = 10, 4 independent experiments, Ifngr1-deficient) or were infected with Arm. (n = 9, 2 independent experiments, control; n = 8, 2 independent experiments, Ifngr1-deficient) or Cl. 13 (n = 15, control; n = 14 (c), n = 15 (d), Ifngr1-deficient) and percentage of Foxp3+ of CD4+ T cells (c), number of Treg cells (d, upper) and CD4+ Tconv cells (d, lower) per spleen, were determined by flow cytometry. e-g, scRNA-seq of splenic Treg cells from uninfected (n = 3) or Cl. 13 infected (n = 5) (d16) Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice. e, Distribution of cells amongst clusters. f, Top 50 ranked significant differentially expressed genes from Treg cells in Foxp3Cre-YFP and Ifngr1L/LFoxp3Cre-YFP mice infected with Cl. 13. g, GSEA overview illustrating pathways upregulated in the control Treg cell gene set compared to Ifngr1-deficient Treg cells during Cl. 13 infection. h, Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice remained uninfected and the expression of T-bet (n = 7), GATA-3 (n = 6, control; n = 5, Ifngr1-deficient) and RORγt (n = 3, control; n = 4, Ifngr1-deficient) by splenic Treg cells were measured by flow cytometry. a-h, Data are presented as mean values and represents biologically independent mice and 2 (a, b, e-h and as indicated) or 3 (c, d) or 5 (as indicated) independent experiments. Statistical significance was determined by an unpaired two-tailed Student’s t-test (b), or by multiple unpaired t-tests (a, c, d, h), relative to Foxp3Cre-YFP mice. Adjusted P values were determined by one-way ANOVA relative to Foxp3Cre-YFP mice (e) or by Wilcoxon rank-sum test (f) or Kolmogorov-Smirnov test (g) relative to infected Foxp3Cre-YFP mice (P values indicated when significant); NS, not significant.

Source data

Extended Data Fig. 4 Intrinsic effects of Ifngr1 deletion from Treg cells using Ifngr1L/LFoxp3Cre-YFPFoxp3Cre-ERT2-GFP heterozygous mice.

a-e, Heterozygous Foxp3Cre-YFP.Foxp3Cre-ERT2-GFP (Cre Het) (n = 5, Uninfected; n = 12, Cl. 13) and Ifngr1L/LFoxp3Cre-YFP.Foxp3Cre-ERT2-GFP (L/L Het) (n = 10, Uninfected; n = 19, Cl. 13) female mice remained uninfected (2 independent experiments) or were infected with Cl. 13 (3 independent experiments). a, Percent expression of GFP+ and YFP+ by splenic CD4+ T cells were determined by flow cytometry. b-e, Expression of the CXCR3 (b), CCR4 (c), CD127 (d) and KLRG1 (e) in splenic Treg cells from indicated mice were measured by flow cytometry. f-h, Heterozygous Foxp3Cre-YFP.Foxp3Cre-ERT2-GFP (Cre Het) and Ifngr1L/LFoxp3Cre-YFP.Foxp3Cre-ERT2-GFP (L/L Het) female mice were infected with Cl. 13. f, Gating strategy for IFNGR1 and IFNGR1+ splenic Treg cells (g, h) from indicated mice. g, h, Expression of T-bet (g) (n = 10, Cre HET; n = 15, L/L HET) and GATA-3 (h) (n = 10, Cre HET; n = 12, L/L HET) by IFNGR1 and IFNGR1+ splenic Treg cells from indicated mice, were measured by flow cytometry. a-h, Data are presented as mean values and represent biologically independent mice and 2 (as indicated) or 3 (a-e, g, h and as indicated) independent experiments. Statistical significance was determined by a two-way AVOVA with multiple comparisons (a, g, h) or by one-way AVOVA with multiple comparisons (b-e) (P values indicated when significant); NS, not significant.

Source data

Extended Data Fig. 5 Treg cells maintain stability during chronic infection.

a, DE of genes induced (upper), or repressed (lower), by Foxp3 in splenic Treg cells of Foxp3Cre-YFP mice compared to Ifngr1L/LFoxp3Cre-YFP mice, that remained uninfected (n = 3) or were infected with Cl. 13 (n = 5) was measured by scRNA-seq. b, c Foxp3Cre-ERT2-GFPRosa26LSL.tdTom mice were treated with tamoxifen for 7d and then remained uninfected or were infected with Cl. 13. b, Experimental schema. c, Representative flow cytometry plot of tdTom and GFP co-expression in splenic CD4+ T cells at steady state (n = 7) or at 16 (n = 5, 2 independent experiments) and 35 (n = 11, 2 independent experiments) dpi with Cl. 13. d-f, Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice were infected with Cl. 13 and levels of Foxp3 (d) (n = 9, control; n = 10, Ifngr1-deficient) and percentage of Nrp1 (e) (n = 9, control; n = 8, Ifngr1-deficient) CD25 (f, left) (n = 11) and level of CD25 (f, right) (n = 11) by splenic Treg cells were determined by flow cytometry. a, c-f, Data are presented as mean values and represent biologically independent mice and 2 (a, d-f and as indicated) or 3 (c) independent experiments. Statistical significance was determined by an unpaired two-tailed Student’s t-test relative to Foxp3Cre-YFP mice (d-f). Adjusted P value was determined by Wilcoxon rank-sum test (a) (P value indicated when significant); NS, not significant.

Source data

Extended Data Fig. 6 IFNγ promotes TH1-like polarization of Treg cells during Influenza A viral infection.

a-d, Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice were infected with IAV. a, b, Percentage of initial body weight over time (a) (numbers of mice used indicated in parentheses) and at day 6 (b) (n = 16, control; n = 17, Ifngr1-deficient). c, d, Expression of T-bet (c) (n = 9, control; n = 7, Ifngr1-deficient) and GATA-3 (d) (n = 10, control; n = 8, Ifngr1-deficient) by lung Treg cells were measured by flow cytometry. a-d. Data are presented as mean values and represent biologically independent mice and 2 (a, c, d) or 3 (b) independent experiments. Statistical significance was determined by two-way ANOVA with multiple comparisons (a) or by an unpaired two-tailed Student’s t-test (b-d), all relative to Foxp3Cre-YFP mice (P values indicated when significant); NS, not significant.

Source data

Extended Data Fig. 7 Ifngr1 deletion from Treg cells does not affect viral load nor markers associated with CD8+ T cell exhaustion during chronic infection.

a, b, Foxp3Cre-YFP, Ifngr1L/L or Ifngr1L/LFoxp3Cre-YFP mice remained uninfected or were infected with Cl. 13. a, Liver histological score from uninfected mice (n = 2) or mice infected with Cl. 13 8 dpi (n = 4, Foxp3Cre-YFP; n = 3, Ifngr1L/L; n = 3, Ifngr1L/LFoxp3Cre-YFP). Score determined as: 1= Triaditis, rare sinusoidal lymphocytes; 2= Triaditis, prominent sinusoidal lymphocytes; 3= Triaditis, prominent sinusoidal lymphocytes, single cell apoptosis. b, Serum AST activity was determined in uninfected mice (n = 9, Foxp3Cre-YFP; n = 2, 1 independent experiment, Ifngr1L/L; n = 8, Ifngr1L/LFoxp3Cre-YFP) or mice infected with Cl. 13, 8 dpi (n = 13, Foxp3Cre-YFP; n = 14, Ifngr1L/LFoxp3Cre-YFP) or 16 dpi (n = 10, Foxp3Cre-YFP; n = 2, 1 independent experiment, Ifngr1L/L; n = 9, Ifngr1L/LFoxp3Cre-YFP). c, Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice were infected with Cl. 13 and the viral load in serum d16 (n = 12, control; n = 11, Ifngr1-deficient; 2 independent experiments) and d30 (n = 11, control; n = 10, Ifngr1-deficient; 2 independent experiments), kidney (n = 7, control; n = 6, Ifngr1-deficient; 1 independent experiment) and liver (n = 5, control; n = 3, Ifngr1-deficient; 1 independent experiment) were determined. d, Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice were infected with Cl. 13 and treated with 200 µg isotype control (n = 3, control; n = 4, Ifngr1-deficient) or anti-PDL1 (n = 5, control; n = 2, Ifngr1-deficient) every 3 d from 26-38 dpi and the viral load in kidney was determined. e, f, Foxp3Cre-YFP (n = 19) or Ifngr1L/LFoxp3Cre-YFP (n = 16) mice were infected with Cl. 13 and IR co-expression (indicated on top) with PD1 (e), and SPICE plots visualizing multiple combinations of IR co-expression (f), by pooled Tetramer+ (GP33, GP276, NP396) splenic CD8+ T cells were determined by flow cytometry. g-j, Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice were infected with Cl. 13 and treated with isotype control (n = 7) or anti-PDL1 (n = 6, control; n = 8, Ifngr1-deficient) as in (d). g, h, Expression of T-bet by bulk splenic CD8+ T cells (g) and PD1 by pooled Tetramer+ (GP33, GP276, NP396) splenic CD8+ T cells (h) were determined by flow cytometry. i, Percentage of pTex (Tcf1+TIM3) and tTex (TCF1TIM3+) splenic CD8+ T cells were determined by flow cytometry. j, Percentage of tTex splenic GP33+CD8+ T cells was determined by flow cytometry. a-j, Data are presented as mean values and represent biologically independent mice and 1 (a, d, and as indicated), 2 (c, g-j) or 3 (b, e, f) independent experiments. a-e, g-j, Statistical significance was determined by a two-way ANOVA with multiple comparisons (a, b) or by an unpaired two-tailed Student’s t-test relative to Foxp3Cre-YFP mice (c, e) or my multiple unpaired Student’s t-tests (d, g-j) (P values indicated when significant); NS, not significant.

Source data

Extended Data Fig. 8 Ifngr1-deficient Treg cells from infected mice maintain suppressive function of naïve CD4+ Tconv cells in vitro and do not impact TH polarization of CD4+ Tconv cells in vivo.

a,b, In vitro microsuppression assay of splenic uninfected CD4+ Tconv (Tresponders) by splenic Treg cells from Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice infected with LCMV Arm. (a) or Cl. 13 (b) (numbers of mice used indicated in parentheses). c, In vitro microsuppression assay of splenic CD44hiCD62Llo CD4+ Tconv (Teff) from Arm. infected Foxp3Cre-YFP mice by splenic Treg cells from Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice infected with LCMV Arm. (numbers of mice used indicated in parentheses). d-g, Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice infected with Cl. 13 and expression of T-bet, GATA-3 and RORγt (d) (n = 8), T-bet and GATA-3 co-expression (e) (n = 8), CXCR3 (f) (n = 8) and CCR4 (g) (n = 8, control; n = 9, Ifngr1-deficient) by splenic CD4+ Tconv cells were measured by flow cytometry. h, Expression of the deleted region of Ifng, normalized to the intact region of Ifng, from IfngL/LFoxp3Cre-YFP mice (n = 4) compared to Foxp3Cre-YFP mice (n = 3). Thy1.2 cells, CD8+ T cells, CD4+ Tconv cells and Treg cells were sorted, gDNA was isolated and qPCR was performed. a-h Data are presented as mean values and represent biologically independent mice and 1 (h), 2 (b-g) or 3 (a) independent experiments. Statistical significance was determined by two-way ANOVA (a-c), or by multiple unpaired Student t-tests (d, e, h), or by an unpaired two-tailed Student’s t-test (f, g), relative to Foxp3Cre-YFP mice (P values indicated when significant); NS, not significant.

Source data

Extended Data Fig. 9 Transcriptomic analysis reveals that TH1-like Treg cells inhibit CD8+ T cell memory during LCMV infection.

a-c, scRNA-seq of splenic pooled Tetramer+ (GP33, GP276, NP396) CD8+ T cells from Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice (n = 3), 16 dpi with Cl. 13. a, UMAP embedding Tetramer+ CD8+ T cells into a two-dimensional space to generate 8 distinct clusters, with categories shown. b, Distribution of cells amongst clusters from individual mice. c, Percentage of cells in the memory cluster. d, e, Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice were infected with Arm. and the expression of effector CD8+ cells (d) (n = 18, control; n = 16, Ifngr1-deficient) and the ratio of MPECs to SLECs within splenic pooled CD44hiCD62Llo Tetramer+ CD8+ T cells (e) (n = 8) were determined by flow cytometry. f, g, Expression of Ifngr1 within the bulk population (f) and within the memory cluster (g) of the splenic pooled Tetramer+ CD8+ T cells from the scRNA-seq dataset in a-c (n = 3). h-j, Rag1–/– mice were reconstituted with Ifngr1–/– (n = 12) or control (WT) (n = 13) Treg cells and infected with Arm. h, i, Frequency of splenic CD44hiCD62Llo (h) and CD44hiCD62Lhi (i, left), and number of CD44hiCD62Lhi (i, right) pooled Tetramer+ CD8+ T cells were determined by flow cytometry. j, Frequency of (left) and total number of (right) MPECs (upper) and SLECs (lower), and the ratio of MPECs to SLECs, splenic pooled Tetramer+ CD8+ T cells were determined by flow cytometry. k, heatmap of DE of the IL16/CD4/CCR5 signaling axis in Treg cells from the scRNA-seq dataset in a-c. l-n, Foxp3Cre-YFP and Ifngr1L/LFoxp3Cre-YFP mice were infected with Arm. and transcripts of the IL16/CD4/CCR5 signaling axis in Treg cells (l) (n = 11) or Il16 (m) (n = 11) and Ccl5 (n) (n = 6, control; n = 9, Ifngr1-deficient) expression in Treg cells and CD44hiCD62Llo CD4+ and CD8+ T cells, were determined by qPCR. a-n, Data are presented as mean values and represent biologically independent mice and 1 (a-c, f, g), 2 (e, k, n) or 3 (d, h-j, l, m) independent experiments. Statistical significance and P values were determined by an unpaired two-tailed Student’s t-test relative to the Foxp3Cre-YFP mice (c-g) or Rag1–/– + WT Treg cells (h-j), or by multiple unpaired Student t-tests relative to Foxp3Cre-YFP mice (l) or by two-way ANOVA with multiple comparisons. (m,n), Adjusted P values were determined by one-way ANOVA (b) or Wilcoxon rank-sum test (k) relative to infected Foxp3Cre-YFP mice (P values indicated when significant); NS, not significant.

Source data

Extended Data Fig. 10 TH1-like Treg cells do not impact tissue resident memory CD8+ T during IAV infection rechallenge.

a-g, Foxp3Cre-YFP or Ifngr1L/LFoxp3Cre-YFP mice were infected with PR8 only (n = 9, control; n = 8, Ifngr1-deficient) or X31 and challenged with PR8 (n = 10, control; n = 9, Ifngr1-deficient). a, Experimental schema. b, Expression of LAP/TGFβ1 by lung Treg cells by percent (upper) and level of expression within LAP/TGFβ1+ Treg cells (lower) were determined by flow cytometry. c, Percentage (upper), and number (lower), of lung IAV pooled Tetramer+ (PA224 and NP366) CD8+ T were determined by flow cytometry. d, Expression of IFNγ by lung CD8+ T cells was measured, after in vitro stimulation with pooled IAV peptides (PA224-233 and NP366-372), by flow cytometry. e, Expression of CXCR3 by lung pooled Tetramer+ CD8+ T cells was measured by flow cytometry. f, Percentage (left), and number (lower), of CD103+CD69+ lung pooled Tetramer+ CD8+ T cells were determined by flow cytometry. g, Blinded histology scores (areas indicated) of paraffin-embedded hematoxylin and eosin stained, lung tissue sections. b-g, Data are presented as mean values and represent biologically independent mice and 3 independent experiments. Statistical significance was determined by multiple unpaired Student’s t-tests relative to Foxp3Cre-YFP mice (P values indicated when significant); NS, not significant.

Source data

Supplementary information

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 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

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gocher-Demske, A.M., Cui, J., Szymczak-Workman, A.L. et al. IFNγ-induction of TH1-like regulatory T cells controls antiviral responses. Nat Immunol 24, 841–854 (2023). https://doi.org/10.1038/s41590-023-01453-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01453-w

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