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:

An immunoregulatory and tissue-residency program modulated by c-MAF in human TH17 cells

A Publisher Correction to this article was published on 17 November 2018

This article has been updated

Abstract

Different types of effector and memory T lymphocytes are induced and maintained in protective or pathological immune responses. Here we characterized two human CD4+ TH17 helper cell subsets that, in the recently activated state, could be distinguished on the basis of their expression of the anti-inflammatory cytokine IL-10. IL-10+ TH17 cells upregulated a variety of genes encoding immunoregulatory molecules, as well as genes whose expression is characteristic of tissue-resident T cells. In contrast, IL-10 TH17 cells maintained a pro-inflammatory gene-expression profile and upregulated the expression of homing receptors that guide recirculation from tissues to blood. Expression of the transcription factor c-MAF was selectively upregulated in IL-10+ TH17 cells, and it was bound to a large set of enhancer-like regions and modulated the immunoregulatory and tissue-residency program. Our results identify c-MAF as a relevant factor that drives two highly divergent post-activation fates of human TH17 cells and provide a framework with which to investigate the role of these cells in physiology and immunopathology.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Transient production of IL-10 is a stable feature of a subset of human memory TH17 cells.
Fig. 2: Production of IL-10 by day 5 (activated) TH17 cells requires chromatin remodeling and upregulation of c-MAF expression.
Fig. 3: TH17-IL-10 and TH17-IL-10+ cells polarize monocytes into M1 and M2 macrophages differentially.
Fig. 4: Distinct transcriptional programs in day 5 (activated) TH17-IL-10+ and TH17-IL-10 cells.
Fig. 5: IL-27 and IL-1β antagonistically promote the expression of genes associated with TH17-IL-10+ cells or TH17-IL-10 cells.
Fig. 6: c-MAF extensively binds bona fide enhancers associated to immune-related genes in TH17-IL-10+ cells.
Fig. 7: c-MAF promotes the immunoregulatory and tissue-residency transcriptional program in day 5 (activated) TH17-IL-10+ cells.

Similar content being viewed by others

Change history

  • 17 November 2018

    In the version of this article initially published, in the legend to Fig. 1b, the description of the frequency of TH17-IL-10+ clones was incomplete for the first group; this should read as follows: “...13 experiments with clones isolated from CCR6+CCR4+CXCR3 T cells...”. Also, the label along the vertical axis of the bottom right plot in Figure 5b was incomplete; the correct label is ‘IFN-γ+ cells (%)’. Finally, in the first sentence of the final paragraph of the final Results subsection, the description of the regions analyzed was incorrect; that sentence should begin: “DNA motif–enrichment analysis of the subset-specific H3K27ac-positive regions...”. The errors have been corrected in the HTML and PDF versions of the article.

References

  1. Sallusto, F., Lenig, D., Förster, R., Lipp, M. & Lanzavecchia, A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401, 708–712 (1999).

    Article  CAS  Google Scholar 

  2. Mueller, S. N. & Mackay, L. K. Tissue-resident memory T cells: local specialists in immune defence. Nat. Rev. Immunol. 16, 79–89 (2016).

    Article  CAS  Google Scholar 

  3. Park, C. O. & Kupper, T. S. The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nat. Med. 21, 688–697 (2015).

    Article  CAS  Google Scholar 

  4. Ouyang, W., Rutz, S., Crellin, N. K., Valdez, P. A. & Hymowitz, S. G. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annu. Rev. Immunol. 29, 71–109 (2011).

    Article  CAS  Google Scholar 

  5. Chen, L. & Flies, D. B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 13, 227–242 (2013).

    Article  Google Scholar 

  6. Attanasio, J. & Wherry, E. J. Costimulatory and coinhibitory receptor pathways in infectious disease. Immunity 44, 1052–1068 (2016).

    Article  CAS  Google Scholar 

  7. Baumeister, S. H., Freeman, G. J., Dranoff, G. & Sharpe, A. H. Coinhibitory pathways in immunotherapy for cancer. Annu. Rev. Immunol. 34, 539–573 (2016).

    Article  CAS  Google Scholar 

  8. Zielinski, C. E. et al. Pathogen-induced human TH17 cells produce IFN-γ or IL-10 and are regulated by IL-1β. Nature 484, 514–518 (2012).

    Article  CAS  Google Scholar 

  9. Saraiva, M. & O’Garra, A. The regulation of IL-10 production by immune cells. Nat. Rev. Immunol. 10, 170–181 (2010).

    Article  CAS  Google Scholar 

  10. Xu, J. et al. c-Maf regulates IL-10 expression during Th17 polarization. J. Immunol. 182, 6226–6236 (2009).

    Article  CAS  Google Scholar 

  11. 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 

  12. 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 

  13. Cao, S., Liu, J., Song, L. & Ma, X. The protooncogene c-Maf is an essential transcription factor for IL-10 gene expression in macrophages. J. Immunol. 174, 3484–3492 (2005).

    Article  CAS  Google Scholar 

  14. Mosser, D. M. & Edwards, J. P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 8, 958–969 (2008).

    Article  CAS  Google Scholar 

  15. Martinez, F. O. & Gordon, S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 6, 13 (2014).

    Article  Google Scholar 

  16. Martinez, F. O., Gordon, S., Locati, M. & Mantovani, A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J. Immunol. 177, 7303–7311 (2006).

    Article  CAS  Google Scholar 

  17. Beyer, M. et al. High-resolution transcriptome of human macrophages. PLoS One 7, e45466 (2012).

    Article  CAS  Google Scholar 

  18. Ranzani, V. et al. The long intergenic noncoding RNA landscape of human lymphocytes highlights the regulation of T cell differentiation by linc-MAF-4. Nat. Immunol. 16, 318–325 (2015).

    Article  CAS  Google Scholar 

  19. Huang, W. et al. DDX5 and its associated lncRNA Rmrp modulate TH17 cell effector functions. Nature 528, 517–522 (2015).

    Article  CAS  Google Scholar 

  20. Evans, H. G. et al. TNF-α blockade induces IL-10 expression in human CD4+ T cells. Nat. Commun. 5, 3199 (2014).

    Article  Google Scholar 

  21. Roychoudhuri, R. et al. BACH2 represses effector programs to stabilize Treg-mediated immune homeostasis. Nature 498, 506–510 (2013).

    Article  CAS  Google Scholar 

  22. Wang, R. et al. Expression of GARP selectively identifies activated human FOXP3+ regulatory T cells. Proc. Natl. Acad. Sci. USA 106, 13439–13444 (2009).

    Article  CAS  Google Scholar 

  23. Hahn, S. A. et al. Soluble GARP has potent antiinflammatory and immunomodulatory impact on human CD4+ T cells. Blood 122, 1182–1191 (2013).

    Article  CAS  Google Scholar 

  24. Probst-Kepper, M. et al. GARP: a key receptor controlling FOXP3 in human regulatory T cells. J. Cell. Mol. Med. 13 9B, 3343–3357 (2009).

    Article  Google Scholar 

  25. Schenk, U. et al. ATP inhibits the generation and function of regulatory T cells through the activation of purinergic P2X receptors. Sci. Signal. 4, ra12 (2011).

    Article  Google Scholar 

  26. Mackay, L. K. et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 352, 459–463 (2016).

    Article  CAS  Google Scholar 

  27. Mackay, L. K. et al. The developmental pathway for CD103+CD8+ tissue-resident memory T cells of skin. Nat. Immunol. 14, 1294–1301 (2013).

    Article  CAS  Google Scholar 

  28. Bromley, S. K., Thomas, S. Y. & Luster, A. D. Chemokine receptor CCR7 guides T cell exit from peripheral tissues and entry into afferent lymphatics. Nat. Immunol. 6, 895–901 (2005).

    Article  CAS  Google Scholar 

  29. Haberman, Y. et al. Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature. J. Clin. Invest. 124, 3617–3633 (2014).

    Article  CAS  Google Scholar 

  30. Barnes, M. G. et al. Gene expression in juvenile arthritis and spondyloarthropathy: pro-angiogenic ELR+ chemokine genes relate to course of arthritis. Rheumatology 43, 973–979 (2004).

    Article  CAS  Google Scholar 

  31. Rao, S. S. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    Article  CAS  Google Scholar 

  32. Heintzman, N. D. et al. Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat. Genet. 39, 311–318 (2007).

    Article  CAS  Google Scholar 

  33. Heintzman, N. D. et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459, 108–112 (2009).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  35. Heinz, S., Romanoski, C. E., Benner, C. & Glass, C. K. The selection and function of cell type-specific enhancers. Nat. Rev. Mol. Cell Biol. 16, 144–154 (2015).

    Article  CAS  Google Scholar 

  36. Kerppola, T. K. & Curran, T. Maf and Nrl can bind to AP-1 sites and form heterodimers with Fos and Jun. Oncogene 9, 675–684 (1994).

    CAS  PubMed  Google Scholar 

  37. Kataoka, K., Noda, M. & Nishizawa, M. Maf nuclear oncoprotein recognizes sequences related to an AP-1 site and forms heterodimers with both Fos and Jun. Mol. Cell. Biol. 14, 700–712 (1994).

    Article  CAS  Google Scholar 

  38. Honda, T. et al. Tuning of antigen sensitivity by T cell receptor-dependent negative feedback controls T cell effector function in inflamed tissues. Immunity 40, 235–247 (2014).

    Article  CAS  Google Scholar 

  39. Kühn, R., Löhler, J., Rennick, D., Rajewsky, K. & Müller, W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 75, 263–274 (1993).

    Article  Google Scholar 

  40. Glocker, E. O. et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N. Engl. J. Med. 361, 2033–2045 (2009).

    Article  CAS  Google Scholar 

  41. Dong, J. et al. IL-10 is excluded from the functional cytokine memory of human CD4+ memory T lymphocytes. J. Immunol. 179, 2389–2396 (2007).

    Article  CAS  Google Scholar 

  42. Gagliani, N. et al. Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation. Nature 523, 221–225 (2015).

    Article  CAS  Google Scholar 

  43. Turner, D. L. & Farber, D. L. Mucosal resident memory CD4 T cells in protection and immunopathology. Front. Immunol. 5, 331 (2014).

    Article  Google Scholar 

  44. Xu, M. et al. c-MAF-dependent regulatory T cells mediate immunological tolerance to a gut pathobiont. Nature 554, 373–377 (2018).

    Article  CAS  Google Scholar 

  45. Samstein, R. M. et al. Foxp3 exploits a pre-existent enhancer landscape for regulatory T cell lineage specification. Cell 151, 153–166 (2012).

    Article  CAS  Google Scholar 

  46. Kwon, H. K., Chen, H. M., Mathis, D. & Benoist, C. Different molecular complexes that mediate transcriptional induction and repression by FoxP3. Nat. Immunol. 18, 1238–1248 (2017).

    Article  CAS  Google Scholar 

  47. Gabryšová, L. et al. c-Maf controls immune responses by regulating disease-specific gene networks and repressing IL-2 in CD4+ T cells. Nat. Immunol. 19, 497–507 (2018).

    Article  Google Scholar 

  48. Förster, R. et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell 99, 23–33 (1999).

    Article  Google Scholar 

  49. El-Behi, M. et al. The encephalitogenicity of TH17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat. Immunol. 12, 568–575 (2011).

    Article  CAS  Google Scholar 

  50. Gaublomme, J. T. et al. Single-cell genomics unveils critical regulators of Th17 cell pathogenicity. Cell 163, 1400–1412 (2015).

    Article  CAS  Google Scholar 

  51. Lee, Y. et al. Induction and molecular signature of pathogenic TH17 cells. Nat. Immunol. 13, 991–999 (2012).

    Article  CAS  Google Scholar 

  52. Ghoreschi, K. et al. Generation of pathogenic TH17 cells in the absence of TGF-β signalling. Nature 467, 967–971 (2010).

    Article  CAS  Google Scholar 

  53. Milner, J. D. et al. Impaired TH17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome. Nature 452, 773–776 (2008).

    Article  CAS  Google Scholar 

  54. Puel, A. et al. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science 332, 65–68 (2011).

    Article  CAS  Google Scholar 

  55. Raffatellu, M. et al. Simian immunodeficiency virus-induced mucosal interleukin-17 deficiency promotes Salmonella dissemination from the gut. Nat. Med. 14, 421–428 (2008).

    Article  CAS  Google Scholar 

  56. Lee, J. S. et al. Interleukin-23-independent IL-17 production regulates intestinal epithelial permeability. Immunity 43, 727–738 (2015).

    Article  CAS  Google Scholar 

  57. Bettelli, E., Korn, T., Oukka, M. & Kuchroo, V. K. Induction and effector functions of TH17 cells. Nature 453, 1051–1057 (2008).

    Article  CAS  Google Scholar 

  58. Kebir, H. et al. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat. Med. 13, 1173–1175 (2007).

    Article  CAS  Google Scholar 

  59. Fujino, S. et al. Increased expression of interleukin 17 in inflammatory bowel disease. Gut 52, 65–70 (2003).

    Article  CAS  Google Scholar 

  60. Kobayashi, T. et al. IL23 differentially regulates the Th1/Th17 balance in ulcerative colitis and Crohn’s disease. Gut 57, 1682–1689 (2008).

    Article  CAS  Google Scholar 

  61. Wilson, N. J. et al. Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat. Immunol. 8, 950–957 (2007).

    Article  CAS  Google Scholar 

  62. Nakae, S., Nambu, A., Sudo, K. & Iwakura, Y. Suppression of immune induction of collagen-induced arthritis in IL-17-deficient mice. J. Immunol. 171, 6173–6177 (2003).

    Article  CAS  Google Scholar 

  63. Patel, D. D. & Kuchroo, V. K. Th17 cell pathway in human immunity: lessons from genetics and therapeutic interventions. Immunity 43, 1040–1051 (2015).

    Article  CAS  Google Scholar 

  64. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  66. 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  Google Scholar 

  67. Blecher-Gonen, R. et al. High-throughput chromatin immunoprecipitation for genome-wide mapping of in vivo protein-DNA interactions and epigenomic states. Nat. Protoc. 8, 539–554 (2013).

    Article  Google Scholar 

  68. Barozzi, I., Termanini, A., Minucci, S. & Natoli, G. Fish the ChIPs: a pipeline for automated genomic annotation of ChIP-Seq data. Biol. Direct. 6, 51 (2011).

    Article  Google Scholar 

  69. Machanick, P. & Bailey, T. L. MEME-ChIP: motif analysis of large DNA datasets. Bioinformatics 27, 1696–1697 (2011).

    Article  CAS  Google Scholar 

  70. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  Google Scholar 

  71. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  Google Scholar 

  72. Mayoral, R. J. & Monticelli, S. Stable overexpression of miRNAs in bone marrow-derived murine mast cells using lentiviral expression vectors. Methods Mol. Biol. 667, 205–214 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Monticelli (Institute for Research in Biomedicine, Bellinzona) for reagents and for critical reading of the manuscript; and I. Barozzi for discussion of ChIP-seq analyses. Supported by the European Research Council (grant 323183 PREDICT to F.S.), the Swiss National Science Foundation (grants 149475 and 170213 to F.S.), the US National Institutes of Health (P01 grant NS076410 to H.L.W.) and the Helmut Horten Foundation (F.S. and the Institute for Research in Biomedicine).

Author information

Authors and Affiliations

Authors

Contributions

D.A. designed and performed experiments, analyzed data and edited the manuscript; M.F. conducted computational data analysis; D.J. performed cell sorting and edited the manuscript; D.H. performed experiments and edited the manuscript; H.L.W., V.K.K. and A.L. provided discussions and edited the manuscript; S.N. designed, performed and supervised experiments, analyzed data and wrote the manuscript; F.S. designed and supervised the study, analyzed data and wrote the manuscript.

Corresponding authors

Correspondence to Samuele Notarbartolo or Federica Sallusto.

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 Sorting strategy and cytokine production by subsets of human memory TH17 cells.

a. Sorting of human memory TH17 cells from PBMCs based on chemokine receptor expression or cytokine secretion. MACS-enriched CD4+ T cells were stained with antibodies to CD45RA, CD25, CCR7, CCR6, CCR4 and CXCR3. CD4+ memory TH17 cells were sorted, after exclusion of CD45RA+CCR7+ naïve T cells (TN) and CD8+ CD14+ CD19+ CD56+ CD25+ cells, as CCR6+CCR4+CXCR3 or, alternatively, as CCR6+CXCR3IL-17A+ (a, lower panels). Dot plots from one donor, representative of more than 10 donors analyzed. b. Cytokine production as assessed by intracellular staining in day 0 (resting) and day 5 (activated) TH17-IL10+ and TH17-IL-10 clone pools after 5 h stimulation with PMA plus ionomycin. Data are represented as mean + 95% c.i., with each dot indicating a T cell clone pool from independent experiments (n ≥ 9) (note: the number of clone pools is paired and the same in TH17-IL-10+ and TH17-IL-10, but can vary for different cytokines (IL-10 and IFN-γ, n = 15; IL-22, n = 13; IL-4, n = 10; GM-CSF, n = 9). *P < 0.05; **P < 0.01; ****P < 0.0001, as determined by ratio paired t test.

Supplementary Figure 2 Gene ontology (GO) analyses of differentially expressed genes and lncRNAs in TH17-IL-10+ and TH17-IL-10 cells.

Gene ontology (GO) analyses on the biological processes associated to the differentially expressed protein-coding genes (a) or lncRNAs (b) in day 0 (resting) and day 5 (activated) TH17-IL-10+ and TH17-IL-10 cells. GO terms are clustered by semantic similarity and a synthetic description is shown. P-value as determined by MetaCore based on hypergeometric distribution; P-value threshold (vertical line) set at 1 × 10–5.

Supplementary Figure 3 Differential surface marker expression in TH17-IL-10+ and TH17-IL-10 cells.

a,b. Expression of CTLA4, PD-1, CD25, CD69, CXCR6, CCR7 and FOXP3 as assessed by flow cytometry in day 5 (activated) TH17-IL-10+ and TH17-IL-10 clone pools. Shown are representative histogram plots (a) and cumulative data of clone pools from independent experiments (mean + 95% c.i.; CTLA-4 and PD-1, n = 10; CD25, CXCR6 and CCR7, n = 9; CD69 and FOXP3, n = 7) (b). *P< 0.05; **P< 0.01; ***P < 0.001; ****P < 0.0001, as determined by ratio paired t test. c. Expansion of TH17-IL-10+ and TH17-IL-10 cells in response to stimulation with CD3/CD28 antibodies. TH17-IL-10+ and TH17-IL-10 cells were stimulated with plate-bound CD3/CD28 antibodies for 48 h and their proliferation was measured at the indicated time points. Shown is the average of two independent experiments (mean ± s.e.m.).

Supplementary Figure 4 Gene set enrichment analysis (GSEA) of TH17-IL-10+ and TH17-IL-10 transcriptional signatures in autoimmune diseases.

Enrichment of TH17-IL-10+ and TH17-IL-10-associated gene signatures in publicly available transcriptional datasets of TH17-mediated autoimmune diseases. Gene set enrichment in ileal biopsies from healthy donors (HD) vs. Crohn’s patients (CD) (a) and in PBMCs and synovial fluid mononuclear cells (SFMCs) from healthy donors (HD) vs. juvenile rheumatoid arthritis patients (RA) (b) are shown. GSEA enrichment results were reported as normalized enrichment score and familywise enrichment (FWER) P value. FWER P values smaller than 0.05 (dashed line) were considered significant. c. c-MAF expression in ileal biopsies from healthy donors (HD), ulcerative colitis (UC) and Crohn’s disease (CD) patients, with histologically graded disease severity, is shown in a box (interquartiles, with a line indicating the median value) and whiskers (min to max values) plot. P-value as determined by Kruskal-Wallis test. Micro. Infl. = microscopic inflammation; Macro. Infl. = macroscopic inflammation.

Supplementary Figure 5 c-MAF binds enhancer-like regions in proximity to genes involved in the immune response.

a. Position weight matrix (PWM) of the top ranked DNA motives identified by the MEME suite software in the day 5 (activated) TH17-IL-10+ c-MAF ChIP-seq dataset; e-values: MAF (DREME), 5.5 × 10–212; MAF (MEME), 6.6 × 10–175; AP-1, 1.7 × 10–146; NFAT, 3.9 × 10–145; RUNX, 2.6 × 10–81; ETS, 1.1 × 10–57; TCF, 1.1 × 10–20. b. Gene Ontology (GO) analysis of genes associated to c-MAF-bound regions in day 5 (activated) TH17-IL-10+ cells. GO biological processes are ranked (top to bottom) according to their binomial P value. c. Graphical representation of a previously characterized (MARE_2) and a novel (IL10_MAF) c-MAF binding sites at the IL10 locus by the Integrative Genome Viewer (IGV). d. The 1 kb genomic region centered on the novel putative IL10 enhancer (IL10_MAF) was aligned to an isometric genomic region centered on a c-MAF peak localized about 9 kb upstream of Il10 in mouse TH17 cells1. The core enhancer region shown is highly conserved between human and mouse (83% identities) and includes a canonical c-MAF binding site (highlighted in yellow). e. The activation status of the newly identified putative IL10 enhancer was evaluated by quantifying H3K27ac and H3K27me3 levels in day 5 (activated) TH17-IL-10+ and TH17-IL-10 cells by ChIP-qPCR (mean + s.e.m.; n = 2). 1Ciofani, M. et al. A validated regulatory network for Th17 cell specification. Cell 151, 289-303 (2012).

Supplementary Figure 6 Examples of c-MAF binding to immunoregulatory and tissue-residency genes loci.

a-g. Graphical representation of c-MAF binding profiles at the locus of the indicated genes using the Integrative Genome Viewer (IGV).

Supplementary Figure 7 c-MAF is partially sufficient to induce IL-10 expression and to repress proinflammatory gene transcription in TH17-IL-10 cells.

a. c-MAF isoform a (upper panels) and isoform b (lower panels) ectopic expression in day 5 (activated) TH17-IL-10 cells as measured by intracellular staining; representative plots (left panels) and combined results (right panels) are displayed. b. IL-10 production secondary to c-MAF ectopic expression in the same cells as determined by intracellular staining. Data are represented as mean + 95% c.i. (isoform a: n = 5, isoform b: n = 7). c. Expression of c-MAF and IL-10 in TH17-IL10 cells upon ectopic expression of c-MAF isoform b. Control (empty vector) cells and c-MAF-b (c-MAF isoform b) expressing TH17-IL10 cells were polyclonally stimulated in the presence or absence of IL-27 (25 ng/ml) and c-MAF and IL-10 expression was measured in day 5 (activated) cells by flow cytometry, after 5 h stimulation with PMA + ionomycin (mean + s.e.m.; n = 3). d. Expression of c-MAF-dependent genes in TH17-IL10 cells upon ectopic expression of c-MAF isoform a (left panel, n = 4) or b (right panel, n = 6), as measured by qPCR. Black bars indicate TH17-IL10+-associated genes, grey bars indicate TH17-IL10-associated genes. Shown is the average log2 fold change over empty vector (mean + s.e.m). n.a., not assessed. e,f. Representative dot plots (e) and cumulative data of expression of IFN-γ and IL-22 (f) as assessed by intracellular staining of Day 5-activated TH17-IL10 cells following ectopic expression of c-MAF isoform b or control empty vector (eV). Data are expressed as percentage over empty vector and represent the mean + s.e.m. (n = 7), with each dot indicating a T cell clone pool from independent experiments. **P < 0.01; ***P < 0.001, as determined by ratio paired t test (a-c) and paired t test (f).

Supplementary Figure 8 Potential cooperative and antagonistic binding of transcription factors in TH17-IL-10+ and TH17-IL-10 cells.

a. c-MAF peaks from day 5 (activated) TH17-IL-10+ cells were ranked according to the fold enrichment over the input, corrected for P-value, and clustered in bins of 1000 peaks (cluster 7 is made of 778 peaks). c-MAF peaks-associated nucleosome-free regions (NFRs) in the same cells were identified from H3K27ac ChIP-seq data and their size was compared to the corresponding NFRs in day 5 (activated) TH17-IL-10 cells. If a NFR was not detected, the size was reported as 0. Each dot in (a) represents a NFR and the white lines indicate median and interquartile ranges. P-values are obtained by Wilcoxon matched-pairs signed rank test. b. Number of total H3K27ac peaks (left panel) identified in day 0 and day 5 TH17-IL-10+ and TH17-IL-10 cells, as assessed by ChIP-seq (MACS P-value ≤ 10 × 10–10, FDR ≤ 5% and fold enrichment ≥ 5). Subset-specific H3K27ac peaks (right panel) were identified, among the total ones, as H3K27ac peaks that were specifically enriched (MACS P-value ≤ 10 × 10–6 and fold enrichment ≥ 3) in the indicated population. Multiple H3K27ac peaks residing within 1 kb were collapsed into a single domain. c. Position weight matrices (PWMs) of DNA enriched motives identified in NFRs associated to recently-activated TH17-IL-10+-specific and TH17-IL-10-specific H3K27ac domains. d. Consensus PWMs of the indicated transcription factors were downloaded from the Hocomoco v.11 database2. Overlapping nucleotides in consensus motifs are highlighted by a light grey box in background e. Gene expression profile of the indicated transcription factors in day 5 TH17-IL-10+ and TH17-IL-10 cells was obtained from RNA-seq data (Fig. 4). 2Kulakovskiy, I.V. et al. HOCOMOCO: towards a complete collection of transcription factor binding models for human and mouse via large-scale ChIP-Seq analysis. Nucleic Acids Res 46, D252-D259 (2018).

Supplementary information

Supplementary Figures and Tables

Supplementary Figures 1-8 and Supplementary Tables 1 and 2

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Aschenbrenner, D., Foglierini, M., Jarrossay, D. et al. An immunoregulatory and tissue-residency program modulated by c-MAF in human TH17 cells. Nat Immunol 19, 1126–1136 (2018). https://doi.org/10.1038/s41590-018-0200-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0200-5

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