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:

Critical role of IRF1 and BATF in forming chromatin landscape during type 1 regulatory cell differentiation

Abstract

Type 1 regulatory T cells (Tr1 cells) are induced by interleukin-27 (IL-27) and have critical roles in the control of autoimmunity and resolution of inflammation. We found that the transcription factors IRF1 and BATF were induced early on after treatment with IL-27 and were required for the differentiation and function of Tr1 cells in vitro and in vivo. Epigenetic and transcriptional analyses revealed that both transcription factors influenced chromatin accessibility and expression of the genes required for Tr1 cell function. IRF1 and BATF deficiencies uniquely altered the chromatin landscape, suggesting that these factors serve a pioneering function during Tr1 cell differentiation.

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: IRF1 and BATF are required for Tr1 differentiation in vitro.
Figure 2: The effects of IRF1 and BATF deficiency on Tr1 function.
Figure 3: IRF1 affects induction and resolution of autoimmune disease.
Figure 4: IRF1 and BATF control generation of IL-10+ Tr1 cells in vivo.
Figure 5: IRF1 and BATF bind to the Il10 locus.
Figure 6: IRF1 and BATF control the Il10 locus.
Figure 7: BATF- and IRF1-deficiences uniquely contribute to Tr1 chromatin and transcriptional landscapes.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Roncarolo, M.G. et al. Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunol. Rev. 212, 28–50 (2006).

    Article  CAS  Google Scholar 

  2. Pot, C., Apetoh, L. & Kuchroo, V.K. Type 1 regulatory T cells (Tr1) in autoimmunity. Semin. Immunol. 23, 202–208 (2011).

    Article  CAS  Google Scholar 

  3. Astier, A.L., Meiffren, G., Freeman, S. & Hafler, D.A. Alterations in CD46-mediated Tr1 regulatory T cells in patients with multiple sclerosis. J. Clin. Invest. 116, 3252–3257 (2006).

    Article  CAS  Google Scholar 

  4. Barrat, F.J. et al. In vitro generation of interleukin 10-producing regulatory CD4(+) T cells is induced by immunosuppressive drugs and inhibited by T helper type 1 (Th1)- and Th2-inducing cytokines. J. Exp. Med. 195, 603–616 (2002).

    Article  CAS  Google Scholar 

  5. Awasthi, A. et al. A dominant function for interleukin 27 in generating interleukin-10-producing anti-inflammatory T cells. Nat. Immunol. 8, 1380–1389 (2007).

    Article  CAS  Google Scholar 

  6. Fitzgerald, D.C. et al. Suppressive effect of IL-27 on encephalitogenic Th17 cells and the effector phase of experimental autoimmune encephalomyelitis. J. Immunol. 179, 3268–3275 (2007).

    Article  CAS  Google Scholar 

  7. Wang, H. et al. IL-27 induces the differentiation of Tr1-like cells from human naive CD4+ T cells via the phosphorylation of STAT1 and STAT3. Immunol. Lett. 136, 21–28 (2011).

    Article  CAS  Google Scholar 

  8. Batten, M. et al. Interleukin 27 limits autoimmune encephalomyelitis by suppressing the development of interleukin 17-producing T cells. Nat. Immunol. 7, 929–936 (2006).

    Article  CAS  Google Scholar 

  9. Apetoh, L. et al. The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat. Immunol. 11, 854–861 (2010).

    Article  CAS  Google Scholar 

  10. Pot, C. et al. Cutting edge: IL-27 induces the transcription factor c-Maf, cytokine IL-21, and the costimulatory receptor ICOS that coordinately act together to promote differentiation of IL-10-producing Tr1 cells. J. Immunol. 183, 797–801 (2009).

    Article  CAS  Google Scholar 

  11. Iwasaki, Y. et al. Egr-2 transcription factor is required for Blimp-1-mediated IL-10 production in IL-27-stimulated CD4+ T cells. Eur. J. Immunol. 43, 1063–1073 (2013).

    Article  CAS  Google Scholar 

  12. Chang, J.T., Wherry, E.J. & Goldrath, A.W. Molecular regulation of effector and memory T cell differentiation. Nat. Immunol. 15, 1104–1115 (2014).

    Article  CAS  Google Scholar 

  13. Josefowicz, S.Z. Regulators of chromatin state and transcription in CD4 T-cell polarization. Immunology 139, 299–308 (2013).

    Article  CAS  Google Scholar 

  14. Tripathi, S.K. & Lahesmaa, R. Transcriptional and epigenetic regulation of T-helper lineage specification. Immunol. Rev. 261, 62–83 (2014).

    Article  CAS  Google Scholar 

  15. Ciofani, M. et al. A validated regulatory network for Th17 cell specification. Cell 151, 289–303 (2012).

    Article  CAS  Google Scholar 

  16. Kurachi, M. et al. The transcription factor BATF operates as an essential differentiation checkpoint in early effector CD8+ T cells. Nat. Immunol. 15, 373–383 (2014).

    Article  CAS  Google Scholar 

  17. Owaki, T. et al. A role for IL-27 in early regulation of Th1 differentiation. J. Immunol. 175, 2191–2200 (2005).

    Article  CAS  Google Scholar 

  18. Pot, C., Apetoh, L., Awasthi, A. & Kuchroo, V.K. Induction of regulatory Tr1 cells and inhibition of T(H)17 cells by IL-27. Semin. Immunol. 23, 438–445 (2011).

    Article  CAS  Google Scholar 

  19. Bettelli, E. et al. Myelin oligodendrocyte glycoprotein-specific T cell receptor transgenic mice develop spontaneous autoimmune optic neuritis. J. Exp. Med. 197, 1073–1081 (2003).

    Article  CAS  Google Scholar 

  20. Schraml, B.U. et al. The AP-1 transcription factor Batf controls T(H)17 differentiation. Nature 460, 405–409 (2009).

    Article  CAS  Google Scholar 

  21. Im, S.H., Hueber, A., Monticelli, S., Kang, K.H. & Rao, A. Chromatin-level regulation of the IL10 gene in T cells. J. Biol. Chem. 279, 46818–46825 (2004).

    Article  CAS  Google Scholar 

  22. Jones, E.A. & Flavell, R.A. Distal enhancer elements transcribe intergenic RNA in the IL-10 family gene cluster. J. Immunol. 175, 7437–7446 (2005).

    Article  CAS  Google Scholar 

  23. Mosser, D.M. & Zhang, X. Interleukin-10: new perspectives on an old cytokine. Immunol. Rev. 226, 205–218 (2008).

    Article  CAS  Google Scholar 

  24. Sjöstrand, M. et al. Expression of the immune regulator tripartite-motif 21 is controlled by IFN regulatory factors. J. Immunol. 191, 3753–3763 (2013).

    Article  Google Scholar 

  25. Yoshida, K. et al. Active repression of IFN regulatory factor-1-mediated transactivation by IFN regulatory factor-4. Int. Immunol. 17, 1463–1471 (2005).

    Article  CAS  Google Scholar 

  26. Ise, W. et al. The transcription factor BATF controls the global regulators of class-switch recombination in both B cells and T cells. Nat. Immunol. 12, 536–543 (2011).

    Article  CAS  Google Scholar 

  27. Hirahara, K. et al. Asymmetric action of STAT transcription factors drives transcriptional outputs and cytokine specificity. Immunity 42, 877–889 (2015).

    Article  CAS  Google Scholar 

  28. Li, P. et al. BATF-JUN is critical for IRF4-mediated transcription in T cells. Nature 490, 543–546 (2012).

    Article  CAS  Google Scholar 

  29. Glasmacher, E. et al. A genomic regulatory element that directs assembly and function of immune-specific AP-1-IRF complexes. Science 338, 975–980 (2012).

    Article  CAS  Google Scholar 

  30. Eason, D.D., Shepherd, A.T. & Blanck, G. Interferon regulatory factor 1 tryptophan 11 to arginine point mutation abolishes DNA binding. Biochim. Biophys. Acta 1446, 140–144 (1999).

    Article  CAS  Google Scholar 

  31. Lee, C.G. et al. A distal cis-regulatory element, CNS-9, controls NFAT1 and IRF4-mediated IL-10 gene activation in T helper cells. Mol. Immunol. 46, 613–621 (2009).

    Article  CAS  Google Scholar 

  32. Buenrostro, J.D., Giresi, P.G., Zaba, L.C., Chang, H.Y. & Greenleaf, W.J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  Google Scholar 

  33. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  Google Scholar 

  34. McCarthy, M.T. & O'Callaghan, C.A. PeaKDEck: a kernel density estimator-based peak calling program for DNaseI-seq data. Bioinformatics 30, 1302–1304 (2014).

    Article  CAS  Google Scholar 

  35. Quinlan, A.R. & Hall, I.M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  Google Scholar 

  36. Anders, S., Pyl, P.T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  Google Scholar 

  37. 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  Google Scholar 

  38. Shishkin, A.A. et al. Simultaneous generation of many RNA-seq libraries in a single reaction. Nat. Methods 12, 323–325 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  40. Kheradpour, P. & Kellis, M. Systematic discovery and characterization of regulatory motifs in ENCODE TF binding experiments. Nucleic Acids Res. 42, 2976–2987 (2014).

    Article  CAS  Google Scholar 

  41. Weirauch, M.T. et al. Determination and inference of eukaryotic transcription factor sequence specificity. Cell 158, 1431–1443 (2014).

    Article  CAS  Google Scholar 

  42. Grant, C.E., Bailey, T.L. & Noble, W.S. FIMO: scanning for occurrences of a given motif. Bioinformatics 27, 1017–1018 (2011).

    Article  CAS  Google Scholar 

  43. Wingender, E., Schoeps, T. & Dönitz, J. TFClass: an expandable hierarchical classification of human transcription factors. Nucleic Acids Res. 41, D165–D170 (2013).

    Article  CAS  Google Scholar 

  44. Blatti, C., Kazemian, M., Wolfe, S., Brodsky, M. & Sinha, S. Integrating motif, DNA accessibility and gene expression data to build regulatory maps in an organism. Nucleic Acids Res. 43, 3998–4012 (2015).

    Article  CAS  Google Scholar 

  45. Watters, A.M., Miraldi, E.R., Martins de Castro, D., Pokrovskii, M. & Bonneau, R. A fast-adapting, scriptable framework for gene regulatory network visualization. Submitted.

  46. Pérez, F.G.B.E. IPython: A System for Interactive Scientific Computing. Comput. Sci. Eng. 9, 21–29 (2007).

    Article  Google Scholar 

  47. Csardi, G. & Nepusz, T. The igraph software package for complex network research. InterJournal, Complex Systems 1695 (2006).

  48. Oliphant, T.E. Python for scientific computing. Comput. Sci. Eng. 9, 10–20 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Kozoriz (Evergrande Center) for cell sorting, F. Quintana (Harvard Medical School) for providing AhR mutant mice, the New York University Langone Medical Center genomics core (A. Heguy and P. Zappile) for help with sequencing, and the Genome Technology Center (at the Laura and Isaac Perlmutter Cancer Center supported by the Cancer Center Support Grant P30CA016087) for help with sequencing. We are most thankful to M. Collins for advice and editing of the manuscript. This work was supported by the US national Institutes of Health (R01 NS 030843, R01 NS 045937, P01 AI073748, P01 NS076410, P01 AI039671 and 5P01 AI056299-10 to V.K.K.; P01NS076410 to D.L. and M.P.; 5T32AI100853 to M.P.; 1R01-DK103358-01 to E.R.M., R.B. and D.L.; and 1R01-GM112192-01 to R.B.), the National Multiple Sclerosis Society (K.K.), Marie Skłodowska-Curie Actions (of the European Commission / Research Executive Agency), grant agreement number 302915, Marie Curie International Outgoing Fellowship (K.K.), the National Science Foundation (IOS-1126971 to R.B.), the DBT-Wellcome Trust (IA/I/12/1/500524 to A.A.), the Klarman Cell Observatory at the Broad Institute (A.R.), the Howard Hughes Medical Institute (A.R.) and the National Cancer Institute (Koch Institute Support (P30-CA14051 to A.R.).

Author information

Authors and Affiliations

Authors

Contributions

K.K. planned, performed and analyzed all of the biological experiments and wrote the manuscript. E.R.M. designed and actuated computational analyses for RNA-seq/ATAC-seq, and contributed to the interpretation of the results and writing of the manuscript. M.P. prepared ATAC-seq libraries, coordinated sequencing with the NYUMC genomics core, and contributed to interpretation of the results and general discussion. A.M., N.Y., X.C., A.W. and N.C. contributed to computational analysis. I.W. prepared RNA-seq libraries and coordinated sequencing. A.A. provided reagents and advice. A.R. developed and undertook transcriptional analysis of IL-27-induced Tr1 cells. R.B. contributed to computational analysis, interpretation of the results and writing of the manuscript. D.L. advised and provided insight into the analysis of ATAC-seq and RNA-seq analysis. V.K.K. supervised the study and wrote the manuscript.

Corresponding author

Correspondence to Vijay K Kuchroo.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 IRF1 and BATF affect T cell gene expression in vitro and in vivo

(a) Quantification of IL-10 and IFN-γ expression in WT, Irf1-/- and Batf-/- naïve cells differentiated in Tr1 conditions for 72 hrs. Dots represent individual mice. (b) FoxP3 expression (mRNA) in WT and Irf1−/− cells treated with TGF-β1 for 72 hrs. (c) Il10 expression in TH0 CD4+ T cells transduced with a retroviral vector expressing IRF1 and/or BATF. (d) Heatmap showing differential gene expression in cells isolated from WT, Irf1−/− and Batf−/− mice immunized with MOG35-55 and restimulated in vitro in the presence of MOG35-55 and IL-23. (e) IL-17A expression measured by FACS in WT and Irf1−/− cells differentiated in vitro in Th17 polarizing conditions (IL-1β+IL-6+IL-23). (f) Quantification of the frequency of IL-17A+ cells shown in (e). (g) Comparison of gene expression in CD4+ T cells differentiated in vitro with IL-27 and CD4+ T cells isolated from mice immunized with MOG35-55 and later restimulated in the presence of IL-27. Shown are fold changes between cells treated with IL-27 and cells treated in the absence of IL-27. Data are pooled from 3 independent experiments (a, b, c), representative of three independent experiments (e, f), pooled from 2 independent experiments (d, g). NS, not significant (P > 0.05); *P < 0.05, **P < 0.001 and ***P < 0.0001 (unpaired t-test, error bars represent mean ±s.e.m.).

Supplementary Figure 2 IRF1 and IRF4 play opposite roles during Tr1 and Th17 differentiation

(a) FACS analysis of IL-10 expression in WT and Irf4−/− cells differentiated in Th0 and Tr1 conditions for 72 hours. (b) Il17a expression (mRNA) in CD4+ T cells differentiated in Th17 polarizing conditions (IL-1β+IL-6+IL-23) with either IRF1 (right bar) or a GFP control (middle bar) retrovirally transduced (c) IL-17A luciferase reporter activity in 293T cells expressing IRF1, IRF4 or both. (d) Il10 expression (mRNA) in CD4+ T cells differentiated in Tr1 polarizing conditions with either IRF4 (right bar) or a GFP control (middle bar) retrovirally transduced. (e) IL-10 luciferase reporter (proximal promoter, HSS+2.98 and CNS-9 fragments) activity in 293T cells expressing IRF1, IRF4 or both. (f) IRF1 binding to sites within Il17a, Il23r and Rorc in WT, Irf1−/− and Irf4−/− CD4+ T cells differentiated in Th17 polarization conditions (IL-1β+IL-6+IL-23). Relative mean enrichment fold (±s.e.m.) in reference to a control antibody is presented. Data are representative of 3 independent experiments (a, c, e), 2 independent experiments (b, d) or pooled from 2 independent experiments (f).

Supplementary Figure 3 Differential accessibility analysis (ATAC-seq)

(a) Number of differentially accessible peaks detected using DESeq2, for many comparisons of interest (log2|FC|>1, FDR=10%). (b) PCA of all ATAC-seq samples, using all ATAC-seq peaks in the dataset (180,478 peaks, log10-transformed intensities).

Supplementary Figure 4 BATF- and IRF1-deficiencies alter Tr1 gene expression patterns

(a) PCA of RNA-seq gene expression data for Irf1−/−, Batf−/− and control Tr1 cells at 72h (15,715 genes, log2-noramlized relative to mean). The PCA scores plot, with samples plotted as functions of gene expression (black (WT), red (Irf1−/−), blue (Batf−/−) balls), is super-imposed on the loadings plot (showing the top 50 genes whose patterns contribute most to the dominant gene expression trends in the dataset). (b) The number of differentially expressed genes, comparing KO to control (FDR=10%, log2|fold-change|>1). (c) The most dramatically KO-dependent genes (FDR=1%, log2|fold-change|>2, for either Batf−/− or Irf1−/−, relative to control).

Supplementary Figure 5 Tr1 IRF1-dependent Network (Zoom-in)

RNA-seq and ATAC-seq datasets were integrated to generate putative transcriptional regulatory networks for Irf1−/− Tr1 cells. Nodes represent differentially expressed transcription factors and target genes, colored according to relative gene expression (log2(Irf1−/−/Control)) at 72h; red and blue indicate high and low relative expression, respectively. Edges are drawn between differentially expressed TFs and putative gene targets if that TF’s motif was enriched in ATAC-seq peaks cis to genes that were increased or decreased in response to Irf1−/− (Praw<10-3, hypergeometric CDF). Edge colors (blue - inhibitory, red - activating) are based on correlation between TF and putative target gene expression. “ON” indicates genes de-repressed in the knockout cells, while “OFF” indicates genes repressed in the knockout cells.

Supplementary Figure 6 Tr1 BATF-dependent Network (Zoom-in)

RNA-seq and ATAC-seq datasets were integrated to generate putative transcriptional regulatory networks for Batf−/− Tr1 cells. Nodes represent differentially expressed transcription factors and target genes, colored according to relative gene expression (log2(Batf−/−/Control)) at 72h; red and blue indicate high and low relative expression, respectively. Edges are drawn between differentially expressed TFs and putative gene targets if that TF’s motif was enriched in ATAC-seq peaks cis to genes that were increased or decreased in response to Batf−/− (Praw<10-3, hypergeometric CDF). Edge colors (blue - inhibitory, red - activating) are based on correlation between TF and putative target gene expression. “ON” indicates genes de-repressed in the knockout cells, while “OFF” indicates genes repressed in the knockout cells.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Table 1 (PDF 1307 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Karwacz, K., Miraldi, E., Pokrovskii, M. et al. Critical role of IRF1 and BATF in forming chromatin landscape during type 1 regulatory cell differentiation. Nat Immunol 18, 412–421 (2017). https://doi.org/10.1038/ni.3683

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3683

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