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:

Epigenetic regulation of the expression of Il12 and Il23 and autoimmune inflammation by the deubiquitinase Trabid

Abstract

The proinflammatory cytokines interleukin 12 (IL-12) and IL-23 connect innate responses and adaptive immune responses and are also involved in autoimmune and inflammatory diseases. Here we describe an epigenetic mechanism for regulation of the genes encoding IL-12 (Il12a and Il12b; collectively called 'Il12' here) and IL-23 (Il23a and Il12b; collectively called 'Il23' here) involving the deubiquitinase Trabid. Deletion of Zranb1 (which encodes Trabid) in dendritic cells inhibited induction of the expression of Il12 and Il23 by Toll-like receptors (TLRs), which impaired the differentiation of inflammatory T cells and protected mice from autoimmune inflammation. Trabid facilitated TLR-induced histone modifications at the promoters of Il12 and Il23, which involved deubiqutination and stabilization of the histone demethylase Jmjd2d. Our findings highlight an epigenetic mechanism for the regulation of Il12 and Il23 and establish Trabid as an innate immunological regulator of inflammatory T cell responses.

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: Zranb1-deficient mice are resistant to CNS inflammation.
Figure 2: Trabid is dispensable in T cells but crucial in DCs for the induction of EAE.
Figure 3: Trabid is required for induction IL-12 and IL-23 (proteins are referred in this case) in cells of the innate immune system.
Figure 4: Trabid is dispensable for TLR-stimulated signaling but is important for the recruitment of c-Rel to Il12 promoters.
Figure 5: Trabid functions as a DUB to facilitate histone demethylation at the Il12b promoter and induction of Il12 expression.
Figure 6: Trabid regulates the stability and function of Jmjd2d.

Similar content being viewed by others

References

  1. Iwasaki, A. & Medzhitov, R. Regulation of adaptive immunity by the innate immune system. Science 327, 291–295 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ganguly, D., Haak, S., Sisirak, V. & Reizis, B. The role of dendritic cells in autoimmunity. Nat. Rev. Immunol. 13, 566–577 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Merad, M., Sathe, P., Helft, J., Miller, J. & Mortha, A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 31, 563–604 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Trinchieri, G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat. Rev. Immunol. 3, 133–146 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Gaffen, S.L., Jain, R., Garg, A.V. & Cua, D.J. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat. Rev. Immunol. 14, 585–600 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Teng, M.W.L. et al. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat. Med. 21, 719–729 (2015).

    Article  CAS  PubMed  Google Scholar 

  7. Langrish, C.L. et al. IL-12 and IL-23: master regulators of innate and adaptive immunity. Immunol. Rev. 202, 96–105 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Sanjabi, S., Hoffmann, A., Liou, H.C., Baltimore, D. & Smale, S.T. Selective requirement for c-Rel during IL-12 P40 gene induction in macrophages. Proc. Natl. Acad. Sci. USA 97, 12705–12710 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hilliard, B.A. et al. Critical roles of c-Rel in autoimmune inflammation and helper T cell differentiation. J. Clin. Invest. 110, 843–850 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Weinmann, A.S. et al. Nucleosome remodeling at the IL-12 p40 promoter is a TLR-dependent, Rel-independent event. Nat. Immunol. 2, 51–57 (2001).

    Article  CAS  PubMed  Google Scholar 

  11. Weinmann, A.S., Plevy, S.E. & Smale, S.T. Rapid and selective remodeling of a positioned nucleosome during the induction of IL-12 p40 transcription. Immunity 11, 665–675 (1999).

    Article  CAS  PubMed  Google Scholar 

  12. Hayes, M.P., Murphy, F.J. & Burd, P.R. Interferon-γ-dependent inducible expression of the human interleukin-12 p35 gene in monocytes initiates from a TATA-containing promoter distinct from the CpG-rich promoter active in Epstein-Barr virus-transformed lymphoblastoid cells. Blood 91, 4645–4651 (1998).

    Article  CAS  PubMed  Google Scholar 

  13. Garrett, S., Fitzgerald, M. & Sullivan, K. LPS and poly I:C induce chromatin modifications at a novel upstream region of the IL-23 p19 promoter. Inflammation 31, 235–246 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Mise-Omata, S. et al. Proximal κB site in the IL-23 p19 promoter is responsible for RelA- and c-Rel-dependent transcription. J. Immunol. 179, 6596–6603 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Bannister, A.J. & Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 21, 381–395 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhu, Y., van Essen, D. & Saccani, S. Cell-type-specific control of enhancer activity by H3K9 Trimethylation. Mol. Cell 46, 408–423 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Evans, P.C. et al. Isolation and characterization of two novel A20-like proteins. Biochem. J. 357, 617–623 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Licchesi, J.D. et al. An ankyrin-repeat ubiquitin-binding domain determines TRABID's specificity for atypical ubiquitin chains. Nat. Struct. Mol. Biol. 19, 62–71 (2012).

    Article  CAS  Google Scholar 

  19. Virdee, S., Ye, Y., Nguyen, D.P., Komander, D. & Chin, J.W. Engineered diubiquitin synthesis reveals Lys29-isopeptide specificity of an OTU deubiquitinase. Nat. Chem. Biol. 6, 750–757 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Tran, H., Hamada, F., Schwarz-Romond, T. & Bienz, M. Trabid, a new positive regulator of Wnt-induced transcription with preference for binding and cleaving K63-linked ubiquitin chains. Genes Dev. 22, 528–542 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Shi, T., Bao, J., Wang, N., Zheng, J. & Wu, D. Identification of small molecule TRABID deubiquitinase inhibitors by computation-based virtual screen. BMC Chem. Biol. 12, 4 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Peterson, L.K. & Fujinami, R.S. Inflammation, demyelination, neurodegeneration and neuroprotection in the pathogenesis of multiple sclerosis. J. Neuroimmunol. 184, 37–44 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Zepp, J., Wu, L. & Li, X. IL-17 receptor signaling and T helper 17-mediated autoimmune demyelinating disease. Trends Immunol. 32, 232–239 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lanzavecchia, A. & Sallusto, F. Regulation of T cell immunity by dendritic cells. Cell 106, 263–266 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Feng, G.J. et al. Extracellular signal-related kinase (ERK) and p38 mitogen-activated protein (MAP) kinases differentially regulate the lipopolysaccharide-mediated induction of inducible nitric oxide synthase and IL-12 in macrophages: Leishmania phosphoglycans subvert macrophage IL-12 production by targeting ERK MAP kinase. J. Immunol. 163, 6403–6412 (1999).

    CAS  PubMed  Google Scholar 

  26. Kim, L. et al. p38 MAPK Autophosphorylation drives macrophage IL-12 production during intracellular infection. J. Immunol. 174, 4178–4184 (2005).

    Article  CAS  PubMed  Google Scholar 

  27. Cho, Y.-C. et al. Enhanced IL-12p40 production in LPS-stimulated macrophages by inhibiting JNK activation by artemisinin. Arch. Pharm. Res. 35, 1961–1968 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Sanjabi, S. et al. A c-Rel subdomain responsible for enhanced DNA-binding affinity and selective gene activation. Genes Dev. 19, 2138–2151 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang, Y. & Reinberg, D. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev. 15, 2343–2360 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Cedar, H. & Bergman, Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat. Rev. Genet. 10, 295–304 (2009).

    Article  CAS  PubMed  Google Scholar 

  31. Snowden, A.W., Gregory, P.D., Case, C.C. & Pabo, C.O. Gene-specific targeting of H3K9 methylation is sufficient for initiating repression in vivo. Curr. Biol. 12, 2159–2166 (2002).

    Article  CAS  PubMed  Google Scholar 

  32. Kim, J. et al. RAD6-mediated transcription-coupled H2B ubiquitylation directly stimulates H3K4 methylation in human cells. Cell 137, 459–471 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cloos, P.A.C., Christensen, J., Agger, K. & Helin, K. Erasing the methyl mark: histone demethylases at the center of cellular differentiation and disease. Genes Dev. 22, 1115–1140 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kroenke, M.A., Carlson, T.J., Andjelkovic, A.V. & Segal, B.M. IL-12- and IL-23-modulated T cells induce distinct types of EAE based on histology, CNS chemokine profile, and response to cytokine inhibition. J. Exp. Med. 205, 1535–1541 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wen, H., Dou, Y., Hogaboam, C.M. & Kunkel, S.L. Epigenetic regulation of dendritic cell-derived interleukin-12 facilitates immunosuppression after a severe innate immune response. Blood 111, 1797–1804 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhou, L. et al. An inducible enhancer required for Il12b promoter activity in an insulated chromatin environment. Mol. Cell. Biol. 27, 2698–2712 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Xu, J. et al. Pioneer factor interactions and unmethylated CpG dinucleotides mark silent tissue-specific enhancers in embryonic stem cells. Proc. Natl. Acad. Sci. USA 104, 12377–12382 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. van Essen, D., Zhu, Y. & Saccani, S. A feed-forward circuit controlling inducible NF-κB target gene activation by promoter histone demethylation. Mol. Cell 39, 750–760 (2010).

    Article  CAS  PubMed  Google Scholar 

  39. Fischle, W., Wang, Y. & Allis, C.D. Histone and chromatin cross-talk. Curr. Opin. Cell Biol. 15, 172–183 (2003).

    Article  CAS  PubMed  Google Scholar 

  40. Fernando, M.D.A., Kounatidis, I. & Ligoxygakis, P. Loss of Trabid, a new negative regulator of the Drosophila immune-deficiency pathway at the level of TAK1, reduces life span. PLoS Genet. 10, e1004117 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Mihaly, S.R., Ninomiya-Tsuji, J. & Morioka, S. TAK1 control of cell death. Cell Death Differ. 21, 1667–1676 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Reiley, W., Zhang, M., Wu, X., Graner, E. & Sun, S.-C. Regulation of the deubiquitinating enzyme CYLD by IκB kinase γ-dependent phosphorylation. Mol. Cell. Biol. 25, 3886–3895 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Jin, W., Zhou, X.F., Yu, J., Cheng, X. & Sun, S.C. Regulation of Th17 cell differentiation and EAE induction by the MAP3K NIK. Blood 113, 6603–6610 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Nelson, J.D., Denisenko, O. & Bomsztyk, K. Protocol for the fast chromatin immunoprecipitation (ChIP) method. Nat. Protoc. 1, 179–185 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Bienz (Medical Research Council, UK) for the plasmid pHM6-HA-Trabid; the EUCOMM Consortium for mice in which Zranb1 was targeted; and personnel from M.D. Anderson Cancer Center resources (flow cytometry, DNA analysis and animal facilities) supported by the National Cancer Institute of the US National Institutes of Health (P30CA016672). Supported by the US National Institutes of Health (AI064639, AI057555 and AI104519), the Center for Inflammation and Cancer at M.D. Anderson Cancer Center, the National Natural Science Foundation of China (81572651), the Thousand Young Talents Plan of China, and the Zhejiang University Special Fund for Fundamental Research.

Author information

Authors and Affiliations

Authors

Contributions

J.J. designed and performed the research, prepared the figures and wrote the manuscript; X.X. made major contributions to the research and prepared some of the figures; Y.X., H.H., Q.Z., and X.C. contributed to experiments; and S.-C.S. supervised the work and wrote the manuscript.

Corresponding author

Correspondence to Shao-Cong Sun.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Zranb1 gene targeting.

(a) Schematic picture of Zranb1 gene targeting using an FRT-LoxP vector, showing the first 6 exons of Zranb1 gene (exons 7-9 are not shown). Targeted mice were crossed with FRT deleter (Rosa26-FLPe) mice to generate Zranb1-floxed mice, which were further crossed with different Cre mice to generate germline KO (CMV-Cre), T-cKO (Cd4-Cre), or DC-cKO (Cd11c-Cre) mice. (b) Genotyping PCR analysis of germline Zranb1 wild-type (WT), KO (KO), and heterozygous (Het) mice using P1/P2 primer pair for wild-type allele and P1/P4 primer pair for KO allele. (c,d) Genotyping PCR of T cell-conditional Zranb1 wild-type (WT, Zranb1+/+Cd4-Cre), T-cKO (Zranb1f/fCd4-Cre), and heterozygous (T-cKO Het, Zranb1+/fCd4-Cre) mice (c) or DC-conditional Zranb1 wild-type (WT, Zranb1+/+Cd11c-Cre), DC-cKO (Zranb1f/fCd11c-Cre), and heterozygous (DC-cKO Het, Zranb1+/fCd11c-Cre) mice (d) using P3/P4 primer pair to amplify WT and floxed alleles and Cre-specific primers for the Cd4-Cre and Cd11c-Cre DNA. (e) RT-PCR was performed to measure the Zranb1 mRNA level in the indicated cell types.

Supplementary Figure 2 Development of cells of the immune system in mice with germline deletion of Zranb1.

(a) Flow cytometry analysis of the percentage (numbers in quadrants) of CD4+CD8+ double-positive (DP), CD4+ and CD8+ single-positive (SP) thymocytes in the thymus (Thy) and B220+ B cells (B) and CD4+ and CD8+ T cells in the spleen (Spl) of wild-type (WT) and KO mice. (b) Flow cytometry analysis of naïve (N, CD62hiCD44lo) and memory (M, CD62loCD44hii) CD4+ T cells and naïve (N), central memory (CM, CD62LhiCD44hi), and effector memory (EM, CD62loCD44hi) CD8+ T cells in the spleen of wild-type and KO mice. (c) Flow cytometry analysis of the Foxp3+ Treg cells in the spleen and thymus of wild-type (WT) or KO mice. (d) Flow cytometry analysis of conventional DC (cDC, CD11c+CD11bhiB220) and plasmacytoid DC (pDC, CD11c+CD11bB220+) percentage in total CD11c-positive cells of Bone marrow (BM) and Spleen (Spl). Data in all panels are presented as a representative FACS plots (left) and mean ± SEM values based on multiple samples (right). Similar results were obtained in three independent experiments. *P<0.05.

Supplementary Figure 3 Trabid is dispensable in T cells for T cell activation and EAE induction.

Age- and sex-matched wild-type (WT) and T-cKO mice were subjected to MOG35-55-induced EAE. (a) Flow cytometry analysis of the CD45+ immune cells (CD4+ and CD8+ T cells and CD11b+ monocytes) infiltrated into the CNS (brain and spinal cord) (n = 3, day 14 post-immunization), presented as a representative plot (left) and summary graph (right). (b) Flow cytometry analysis of the IFN-γ-producing TH1 and IL-17A-producing TH17 cells in the CNS (percent of CD4+CD45+ cells) and draining lymph nodes (percent of CD4+ T cells) (n = 3, day 14 post-immunization). Data are presented as a representative plot (left, CNS only) and summary graph of absolute cell numbers (right). (c) ELISA analysis of the indicated cytokines in the supernatants of cultures of splenocytes isolated from WT and T-cKO EAE mice (day 14 after immunization) and restimulated in vitro with MOG peptide (20 μg/ml for 48 h). (d) Flow cytometry analysis of the frequency of TH1, TH17, and TREG cells in wild-type and T-cKO naive CD4+ T cells, stimulated for 4 days with plate-bound anti-CD3 and anti-CD28 antibodies under TH0, TH1, TH17, or TREG conditions. (e) Flow cytometry analysis of cell proliferation, based on CFSE dilution, of naïve T cells stimulated with plate-bound anti-CD3 (5 μg/mL) and anti-CD28 (1 μg/mL) for 48 h. (f) ELISA of IFN-γ and IL-2 using supernatants of the cell cultures described in e. Data are representative of three independent experiments. Error bars are mean ± SEM values.

Supplementary Figure 4 Effect of Trabid deficiency on the function of DCs in regulating T cell differentiation.

(a,b) Flow cytometry analysis of Foxp3+ TREG cells (a) or IFN-γ+ TH1 and IL-17+ TH17 cells (b) in wild-type naïve CD4+ T cells stimulated for 4 days with plate-bound anti-CD3 and anti-CD28 plus supernatant of LPS-activated wild-type (WT) or DC-cKO BMDCs (LPS-DC) in the absence (–) or presence (+) of the indicated cytokines.

Supplementary Figure 5 Trabid regulates LPS-stimulated c-Rel recruitment and histone modifications at the promoters of Il12a and Il12b.

(a) ChIP assays of c-Rel recruitment to the promoter of the indicated genes using wild-type (WT) and DC-cKO BMDCs that were either unstimulated (0 h) or stimulated for 6 h with LPS. Data are presented as percentage of the total input DNA, as determined by QPCR assays. (b) Luciferase assays using HEK293 cells transfected with an Il12b-luciferase reporter plasmid in the presence (+) or absence (–) of an empty vector (Vector) or expression vectors encoding c-Rel, Trabid, or a catalytically inactive Trabid mutant, C443 (Mut Trabid). Luciferase activity is presented as fold based on empty vector. (c) ChIP analyses of H3K9 dimethylation (H3K9me2) and trimethylation (H3K9me3) in the Il12a promoter using wild-type or DC-cKO BMDCs that were untreated (NT) or stimulated with LPS for 6 h. The Y axis is percentage (%) of total H3. Data are presented as mean ± SEM values and representative of at least three independent experiments. Statistical analyses represent variations in technical replicates *P < 0.05; **P < 0.01.

Supplementary Figure 6 Trabid deficiency suppresses the recruitment of Pol II complex components to the Il12b promoter.

ChIP analysis of the recruitment of Pol II, Pol II pS5, TFIIB, and TFIID to the indicated promoters in wild-type (WT) or DC-cKO BMDCs. The Y axis is percentage (%) of the DNA bound to the specific factors based on total input DNA. Data are presented as mean ± SEM values and representative of at least three independent experiments. Statistical analyses represent variations in technical replicates. *P < 0.05; **P < 0.01.

Supplementary Figure 7 Jmjd2d is regulated by Trabid and mediates induction of Il12 genes.

(a,b) ChIP analysis of the binding of indicated Jmjd2 family members to different regions of Il12b locus in wild-type (WT) and DC-cKO BMDCs (a) or wild-type and Rel-KO BMDCs (b) that were either not treated (NT) or stimulated with LPS for 6 h. (c) Immunoblotting analysis of Jmjd2d in wild-type BMDCs transduced with pGIPZ lentiviral vectors encoding a non-silencing control shRNA (C) or two different Kdm4d-specific shRNAs. The cells were stimulated with LPS for 6 h to induce the level of Jmjd2d. (d) qRT-PCR analysis of the indicated genes in LPS-stimulated BMDCs described in c. Data are presented as fold relative to the Actb mRNA level. (e) Immunoblotting analysis of HA-tagged Trabid (or Trabid C443A) and FLAG-tagged Jmjd2d in anti-FLAG (Jmjd2d) immunoprecipitates (top two panels) or lysates (lower panels) of HEK293 cells transfected with the indicated plasmids. (f,g) Immunoblotting analysis of Jmjd2d (f) and qRT-PCR analysis of the indicated genes in DC-cKO BMDCs reconstituted with pCLXSN(GFP) (Vec) or pCLXSN(GFP)-HA-Jmjd2d (Jmjd2d). Data are presented as mean ± SEM values and representative of at least three independent experiments. *P < 0.05; **P<0.01.

Supplementary Figure 8 A model of Trabid function in TLR-mediated induction of Il12b.

LPS induces the expression of the Jmjd2d gene (Kdm4d) via an unknown signaling pathway. Jmjd2d mediates removal of H3K9me2 and H3K9me3 from the Il12b promoter, thereby promoting c-Rel binding to the κB element and induction of Il12b expression. The fate of Jmjd2d in TLR-stimulated DCs is tightly controlled by ubiquitination. An unknown E3 ubiquitin ligase mediates Jmjd2d ubiquitination and degradation, whereas Trabid deubiquitinates Jmjd2d to prevent its degradation. Trabid deficiency causes elevated ubiquitination and degradation of Jmjd2d and, thus, reduces the level of Il12b induction. Similar mechanisms may apply to the induction of Il12a and Il23a genes.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1–3 (PDF 2324 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jin, J., Xie, X., Xiao, Y. et al. Epigenetic regulation of the expression of Il12 and Il23 and autoimmune inflammation by the deubiquitinase Trabid. Nat Immunol 17, 259–268 (2016). https://doi.org/10.1038/ni.3347

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3347

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