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:

Jmjd1c demethylates STAT3 to restrain plasma cell differentiation and rheumatoid arthritis

Abstract

Appropriate regulation of B cell differentiation into plasma cells is essential for humoral immunity while preventing antibody-mediated autoimmunity; however, the underlying mechanisms, especially those with pathological consequences, remain unclear. Here, we found that the expression of Jmjd1c, a member of JmjC domain histone demethylase, in B cells but not in other immune cells, protected mice from rheumatoid arthritis (RA). In humans with RA, JMJD1C expression levels in B cells were negatively associated with plasma cell frequency and disease severity. Mechanistically, Jmjd1c demethylated STAT3, rather than histone substrate, to restrain plasma cell differentiation. STAT3 Lys140 hypermethylation caused by Jmjd1c deletion inhibited the interaction with phosphatase Ptpn6 and resulted in abnormally sustained STAT3 phosphorylation and activity, which in turn promoted plasma cell generation. Germinal center B cells devoid of Jmjd1c also acquired strikingly increased propensity to differentiate into plasma cells. STAT3 Lys140Arg point mutation completely abrogated the effect caused by Jmjd1c loss. Mice with Jmjd1c overexpression in B cells exhibited opposite phenotypes to Jmjd1c-deficient mice. Overall, our study revealed Jmjd1c as a critical regulator of plasma cell differentiation and RA and also highlighted the importance of demethylation modification for STAT3 in B cells.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: B cell-expressed Jmjd1c suppresses autoimmune arthritis.
Fig. 2: Jmjd1c restrains STAT3 activation in B cells.
Fig. 3: Jmjd1c deficiency enhances B cell differentiation into plasma cells.
Fig. 4: STAT3 hyperactivation accounts for the enhanced plasma cell differentiation in Jmjd1c-KO B cells.
Fig. 5: Jmjd1c demethylates STAT3 to facilitate its dephosphorylation by Ptpn6.
Fig. 6: Jmjd1c demethylates STAT3 at Lys140.
Fig. 7: Jmjd1c overexpression represses plasma cell differentiation and rheumatoid arthritis progression.

Similar content being viewed by others

Data availability

RNA-seq data and CUT&Tag data reported in this paper are accessible at the Gene Expression Omnibus under accessions GSE193752 and GSE195462 respectively. MS data are available at iProX (PXD031855). There are no restrictions for data availability. Source data are provided with this paper.

Code availability

The codes for RNA-seq and CUT&Tag data analyses reported in this study have been deposited in GitHub at https://github.com/MikeYuliang/NGSdataanalysis/.

Any additional information required to reanalyze the data is available from the corresponding author upon request.

References

  1. Nutt, S. L., Hodgkin, P. D., Tarlinton, D. M. & Corcoran, L. M. The generation of antibody-secreting plasma cells. Nat. Rev. Immunol. 15, 160–171 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Ise, W. & Kurosaki, T. Plasma cell differentiation during the germinal center reaction. Immunol. Rev. 288, 64–74 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Young, C. & Brink, R. The unique biology of germinal center B cells. Immunity 54, 1652–1664 (2021).

    Article  CAS  PubMed  Google Scholar 

  4. Cyster, J. G. & Allen, C. D. C. B cell responses: cell interaction dynamics and decisions. Cell 177, 524–540 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Mesin, L., Ersching, J. & Victora, G. D. Germinal center B cell dynamics. Immunity 45, 471–482 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Victora, G. D. & Nussenzweig, M. C. Germinal centers. Annu. Rev. Immunol. 30, 429–457 (2012).

    Article  CAS  PubMed  Google Scholar 

  7. Elsner, R. A. & Shlomchik, M. J. Germinal center and extrafollicular B cell responses in vaccination, immunity, and autoimmunity. Immunity 53, 1136–1150 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Minnich, M. et al. Multifunctional role of the transcription factor Blimp-1 in coordinating plasma cell differentiation. Nat. Immunol. 17, 331–343 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Malkiel, S., Barlev, A. N., Atisha-Fregoso, Y., Suurmond, J. & Diamond, B. Plasma cell differentiation pathways in systemic lupus erythematosus. Front. Immunol. 9, 427 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. McInnes, I. B. & Schett, G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 365, 2205–2219 (2011).

    Article  CAS  PubMed  Google Scholar 

  11. Dörner, T., Giesecke, C. & Lipsky, P. E. Mechanisms of B cell autoimmunity in SLE. Arthritis Res. Ther. 13, 243 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Winter, O., Dame, C., Jundt, F. & Hiepe, F. Pathogenic long-lived plasma cells and their survival niches in autoimmunity, malignancy, and allergy. J. Immunol. 189, 5105–5111 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. William, J., Euler, C. & Shlomchik, M. J. Short-lived plasmablasts dominate the early spontaneous rheumatoid factor response: differentiation pathways, hypermutating cell types, and affinity maturation outside the germinal center. J. Immunol. 174, 6879–6887 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Eisenberg, R. & Albert, D. B cell-targeted therapies in rheumatoid arthritis and systemic lupus erythematosus. Nat. Clin. Pract. Rheumatol. 2, 20–27 (2006).

    Article  CAS  PubMed  Google Scholar 

  15. Edwards, J. C. & Cambridge, G. B-cell targeting in rheumatoid arthritis and other autoimmune diseases. Nat. Rev. Immunol. 6, 394–403 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Townsend, M. J., Monroe, J. G. & Chan, A. C. B cell-targeted therapies in human autoimmune diseases: an updated perspective. Immunol. Rev. 237, 264–283 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Zhu, N. et al. MLL-AF9- and HOXA9-mediated acute myeloid leukemia stem cell self-renewal requires JMJD1C. J. Clin. Invest. 126, 997–1011 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Peeken, J. C. et al. Epigenetic regulation of NFE2 overexpression in myeloproliferative neoplasms. Blood 131, 2065–2073 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kuroki, S. et al. JMJD1C, a JmjC domain-containing protein, is required for long-term maintenance of male germ cells in mice1. Biol. Reprod. 89, 93 (2013).

    Article  PubMed  CAS  Google Scholar 

  20. Viscarra, J. A., Wang, Y., Nguyen, H. P., Choi, Y. G. & Sul, H. S. Histone demethylase JMJD1C is phosphorylated by mTOR to activate de novo lipogenesis. Nat. Commun. 11, 796 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Thompson, S. D. et al. Genome-wide association analysis of juvenile idiopathic arthritis identifies a new susceptibility locus at chromosomal region 3q13. Arthritis Rheum. 64, 2781–2791 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Syed, A. A. S., He, L. & Shi, Y. The potential effect of aberrant testosterone levels on common diseases: a mendelian randomization study. Genes 11, 721 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  23. Reljic, R., Wagner, S. D., Peakman, L. J. & Fearon, D. T. Suppression of signal transducer and activator of transcription 3-dependent B lymphocyte terminal differentiation by BCL-6. J. Exp. Med. 192, 1841–1848 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Diehl, S. A. et al. STAT3-mediated up-regulation of BLIMP1 Is coordinated with BCL6 down-regulation to control human plasma cell differentiation. J. Immunol. 180, 4805–4815 (2008).

    Article  CAS  PubMed  Google Scholar 

  25. Fornek, J. L. et al. Critical role for Stat3 in T-dependent terminal differentiation of IgG B cells. Blood 107, 1085–1091 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kwon, H. et al. Analysis of interleukin-21-induced Prdm1 gene regulation reveals functional cooperation of STAT3 and IRF4 transcription factors. Immunity 31, 941–952 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Haga, S. et al. Stat3 protects against Fas-induced liver injury by redox-dependent and -independent mechanisms. J. Clin. Invest. 112, 989–998 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bromberg, J. F. et al. Stat3 as an oncogene. Cell 98, 295–303 (1999).

    Article  CAS  PubMed  Google Scholar 

  29. Oike, T. et al. Stat3 as a potential therapeutic target for rheumatoid arthritis. Sci. Rep. 7, 10965 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Miklossy, G., Hilliard, T. S. & Turkson, J. Therapeutic modulators of STAT signalling for human diseases. Nat. Rev. Drug. Discov. 12, 611–629 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kim, S. M. et al. Regulation of mouse steroidogenesis by WHISTLE and JMJD1C through histone methylation balance. Nucleic Acids Res. 38, 6389–6403 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kim, E. et al. Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells. Cancer Cell 23, 839–852 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dasgupta, M., Dermawan, J. K., Willard, B. & Stark, G. R. STAT3-driven transcription depends upon the dimethylation of K49 by EZH2. Proc. Natl Acad. Sci. USA 112, 3985–3990 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kim, M., Morales, L. D., Jang, I.-S., Cho, Y.-Y. & Kim, D. J. Protein tyrosine phosphatases as potential regulators of STAT3 signaling. Int. J. Mol. Sci. 19, 2708 (2018).

    Article  PubMed Central  CAS  Google Scholar 

  35. Chen, K. et al. Methyltransferase SETD2-mediated methylation of STAT1 is critical for interferon antiviral activity. Cell 170, 492–506 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Yang, J. et al. Reversible methylation of promoter-bound STAT3 by histone-modifying enzymes. Proc. Natl Acad. Sci. USA 107, 21499–21504 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu, L., McBride, K. M. & Reich, N. C. STAT3 nuclear import is independent of tyrosine phosphorylation and mediated by importin-alpha3. Proc. Natl Acad. Sci. USA 102, 8150–8155 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zouein, F. A., Duhé, R. J. & Booz, G. W. JAKs go nuclear: emerging role of nuclear JAK1 and JAK2 in gene expression and cell growth. Growth Factors 29, 245–252 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Watanabe, S. et al. JMJD1C demethylates MDC1 to regulate the RNF8 and BRCA1-mediated chromatin response to DNA breaks. Nat. Struct. Mol. Biol. 20, 1425–1433 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Milner, J. D. et al. Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood 125, 591–599 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Flanagan, S. E. et al. Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat. Genet. 46, 812–814 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Garbers, C., Heink, S., Korn, T. & Rose-John, S. Interleukin-6: designing specific therapeutics for a complex cytokine. Nat. Rev. Drug. Discov. 17, 395–412 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Kang, S., Tanaka, T., Narazaki, M. & Kishimoto, T. Targeting Interleukin-6 signaling in clinic. Immunity 50, 1007–1023 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Ohshima, S. et al. Interleukin 6 plays a key role in the development of antigen-induced arthritis. Proc. Natl Acad. Sci. USA 95, 8222–8226 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Alonzi, T. et al. Interleukin 6 is required for the development of collagen-induced arthritis. J. Exp. Med. 187, 461–468 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ciobanu, D. A. et al. JAK/STAT pathway in pathology of rheumatoid arthritis. Exp. Ther. Med. 20, 3498–3503 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kishimoto, T. IL-6: from its discovery to clinical applications. Int. Immunol. 22, 347–352 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Anderson, A. E. et al. Expression of STAT3-regulated genes in circulating CD4+ T cells discriminates rheumatoid arthritis independently of clinical parameters in early arthritis. Rheumatology 58, 1250–1258 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Krause, A., Scaletta, N., Ji, J. D. & Ivashkiv, L. B. Rheumatoid arthritis synoviocyte survival is dependent on Stat3. J. Immunol. 169, 6610–6616 (2002).

    Article  CAS  PubMed  Google Scholar 

  50. O’Shea, J. J., Gadina, M. & Schreiber, R. D. Cytokine signaling in 2002: new surprises in the Jak–Stat pathway. Cell 109, S121–131 (2002).

    Article  PubMed  Google Scholar 

  51. Barnden, M. J., Allison, J., Heath, W. R. & Carbone, F. R. Defective TCR expression in transgenic mice constructed using cDNA-based alpha- and beta-chain genes under the control of heterologous regulatory elements. Immunol. Cell Biol. 76, 34–40 (1998).

    Article  CAS  PubMed  Google Scholar 

  52. Shih, T. A., Roederer, M. & Nussenzweig, M. C. Role of antigen receptor affinity in T cell-independent antibody responses in vivo. Nat. Immunol. 3, 399–406 (2002).

    Article  CAS  PubMed  Google Scholar 

  53. Aletaha, D. et al. 2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 62, 2569–2581 (2010).

    Article  PubMed  Google Scholar 

  54. Prevoo, M. L. et al. Modified disease activity scores that include twenty-eight-joint counts. Development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum. 38, 44–48 (1995).

    Article  CAS  PubMed  Google Scholar 

  55. Aletaha, D. & Smolen, J. S. The Simplified Disease Activity Index (SDAI) and Clinical Disease Activity Index (CDAI) to monitor patients in standard clinical care. Best. Pract. Res. Clin. Rheumatol. 21, 663–675 (2007).

    Article  PubMed  Google Scholar 

  56. Klarquist, J. & Janssen, E. M. The bm12 inducible model of systemic lupus erythematosus in C57BL/6 mice. J. Vis. Exp. 105, e53319 (2015).

    Google Scholar 

  57. Luo, D. et al. Deltex2 represses MyoD expression and inhibits myogenic differentiation by acting as a negative regulator of Jmjd1c. Proc. Natl Acad. Sci. USA 114, E3071–e3080 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Chen, J. et al. Methyltransferase Nsd2 ensures germinal center selection by promoting adhesive interactions between B cells and follicular dendritic cells. Cell Rep. 25, 3393–3404 (2018).

    Article  CAS  PubMed  Google Scholar 

  59. Long, X. et al. Histone methyltransferase Nsd2 is required for follicular helper T cell differentiation. J. Exp. Med. 217, e20190832 (2020).

    Article  PubMed  CAS  Google Scholar 

  60. Ma, K. et al. TLR4+CXCR4+ plasma cells drive nephritis development in systemic lupus erythematosus. Ann. Rheum. Dis. 77, 1498–1506 (2018).

    Article  CAS  PubMed  Google Scholar 

  61. Hasegawa, T. et al. Identification of a novel arthritis-associated osteoclast precursor macrophage regulated by FoxM1. Nat. Immunol. 20, 1631–1643 (2019).

    Article  CAS  PubMed  Google Scholar 

  62. Tanaka, S. et al. Tet2 and Tet3 in B cells are required to repress CD86 and prevent. autoimmunity. Nat. Immunol. 21, 950–961 (2020).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank all the members of the Wang laboratory for feedback. We thank W. Zhang, W. Zeng and J. Li for genetic mouse generation and technical support. This study was supported by grants from the National Natural Science Foundation of China (82101827 to J.C., 31970828 to X.W. and 82171793 to N.C.), National Key R&D Program of China (2018YFC1003900 to X.W.), China Postdoctoral Science Foundation (2020M681665 to J.C.) and Jiangsu Outstanding Young Investigator Program (BK20200030 to X.W.).

Author information

Authors and Affiliations

Authors

Contributions

X.W. and J.C. conceived and directed the study. Y.Y., X.Y., S.W. and Y.C. designed and performed most of the experiments. N.C. designed and directed the experiments with human samples. Y.W. performed the bioinformatic analysis. H.Z., X.L., X.D. and S.Z. helped with mouse caring and some in vitro experiments. X.W. and J.C. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Nan Che, Jingjing Chen or Xiaoming Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Stephen Nutt and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey, in collaboration with the Nature Immunology team. Peer reviewer reports are available.

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 Normal B cell development in the absence of Jmjd1c.

a, The Jmjd1c gene conditional targeting strategy. Exon 9 and 10 was floxed and deleted by crossing to Cre+ mouse strain. Enzymatic JmjC domain was encoded by Exon 21-26. b, Left, flow cytometric analysis showing expression of CD11b, B220, CD43, BP-1, CD24, IgM and IgD in bone marrow cells from Jmjd1c+/+ Mb1Cre+ (n=4) and Jmjd1cfl/fl Mb1Cre+ mice (n=4). Right, absolute numbers of B220+CD43+ and B220+CD43 bone marrow cells per femur in each developmental stage. c,Left, expression of B220, IgD, IgM, AA4.1, CD23, and CD21 in splenocytes from Jmjd1c+/+ Mb1Cre+ (n=5) and Jmjd1cfl/fl Mb1Cre+ (n=5) mice. Right, absolute number of B220+ splenocytes in each developmental stage. Data are means ± SEM. Student’s t tests were used (NS, not significant).

Source data

Extended Data Fig. 2 Expression of Jmjd1c during B cell differentiation.

a, mRNA of Jmjd1c were examined by RT-qPCR in cells as indicated. b, Jmjd1c protein expression was examined by immunoblot with cells as indicated. Naïve B cells (NB) were sorted as CD19+IgDhi and stimulated with anti-IgM plus anti-CD40 for 24hr to obtain activated B cells (ABC). Germinal center B cells (GCB, CD19+IgDlowFas+GL7+) and plasma cells (PC, B220lowCD138+) were sorted from day 8 SRBC immunized mice.

Source data

Extended Data Fig. 3 Jmjd1c expressed in B cells was dispensable for T cell helper function, but was required for antibody production.

a, Left, representative flow cytometry plots of splenic CD4+ T cell subsets: CD4+ naïve T (CD44-CD62L+) and CD4+ T effector memory (CD44+CD62L) cells from Jmjd1c+/+ Mb1Cre+ (n=4) and Jmjd1cfl/fl Mb1Cre+ mice (n=5). Right, frequency of the T cell populations. b, Splenocytes from Jmjd1c+/+ Mb1Cre+ and Jmjd1cfl/fl Mb1Cre+ mice were cultured with or without 1 μg/ml synthetic Eα52–68 peptide for 1 h, and then the levels MHC-II-Ea complex on B cells were measured with Y-Ae antibody by FACS. c and d, Jmjd1c+/+ Mb1Cre+ and Jmjd1cfl/fl Mb1Cre+ mice were induced for the collagen arthritis. (C) Left, representative flow cytometry plots of Th1 (CD4+IFN-γ+) cells from inguinal lymph nodes. Right, number of the Th1 cells. (D) ELISA of anti- collagen IgG2b (left), IgG2c (middle) and IgG1 (right) from serial serum samples. Data are means ± SEM. Student’s t tests were used for analyses in (a) and (c) (NS, not significant). Two-way ANOVA were used for (d) (***p < 0.001). All data are representative of 3 individual experiments.

Source data

Extended Data Fig. 4 Increased antibody production in Jmjd1cfl/fl Mb1Cre+ mice.

a, ELISA of NP-specific IgM and IgG antibody from serum of Jmjd1c+/+ Mb1Cre+ and Jmjd1cfl/fl Mb1Cre+ mice immunized with NP-Ficoll for 7 days. These data are representative of 2 individual experiments. b, ELISA of NP-specific IgG1 antibody from serum of Jmjd1c+/+ Mb1Cre+ and Jmjd1cfl/fl Mb1Cre+ mice immunized with NP-KLH at day 7, 14, 21. These data are representative of 3 individual experiments. Data are means ± SEM. Two-way ANOVA were used (***p < 0.001).

Source data

Extended Data Fig. 5 Increased Stat3 phosphorylation and antibody production in Jmjd1cfl/fl CD19Cre+ mice.

a, B cells from Jmjd1c+/+ CD19Cre+ and Jmjd1cfl/fl CD19Cre+ mice were stimulated with anti-IgM (left) or IL-6 (right) for 16 h. Expression of p-Stat3 was subsequently analyzed by immunoblot. Blots were probed against total Stat3, phosphorylated Stat3 (Y705) and β-actin. b, ELISA of NP-specific IgM and IgG from serum of Jmjd1c+/+ CD19Cre+ and Jmjd1cfl/fl CD19Cre+ mice immunized with NP-Ficoll for 7 days. c, ELISA of NP-specific IgG1 from serum of Jmjd1c+/+ CD19Cre+ and Jmjd1cfl/fl CD19Cre+ mice immunized with NP-KLH at day 7, 14, 21. Data are means ± SEM. Two-way ANOVA were used (***p < 0.001). Data are representative of 2 individual experiments.

Source data

Extended Data Fig. 6 The vector map of MSCV-U6-sgRNA-Thy1.1.

The U6 promoter and gRNA scaffold were synthesized and cloned into the MSCV-Thy1.1 retroviral plasmid to generate the MSCV-U6-sgRNA-Thy1.1 vector.

Extended Data Fig. 7 Jmjd1c did not regulate H3K9 methylation in B cells.

a, B cells were sorted from Jmjd1c+/+ Mb1Cre+ and Jmjd1cfl/fl Mb1Cre+ mice. The level of Jmjd1c and H3K9me was subsequently analyzed by western blot. Blots were probed against Jmjd1c, β-actin, H3K9me1, H3K9me2, H3K9me3 and total H3. Data are representative of 2 individual experiments. b, Anti-H3K9me2 CUT&Tag was performed with B cells from Jmjd1c+/+ Mb1Cre+ and Jmjd1cfl/fl Mb1Cre+ mice to analyze the genomic distribution of H3K9me2 modification. The overall H3K9me2 peak signal was shown in averaged profile plot (left) or heatmap (right).

Source data

Extended Data Fig. 8 Stat3 interacted with Jmjd1c.

a, IB analysis of whole-cell lysates (WCLs) and immunoprecipitation (IP) of 293T cells transfected with the indicated plasmids. b, Schematic of Flag-tagged Jmjd1c deletions used to identify the Stat3-interacting domain. Immunoblot analysis of WCLs and IP of 293T cells transfected with the indicated expression vectors. Data are representative of 2 individual experiments.

Source data

Extended Data Fig. 9 Jmjd1c expression in B cells suppressed lupus-like disease.

a and b, Jmjd1c+/+ Mb1Cre+, Jmjd1cfl/fl Mb1Cre+ and Jmjd1cOEMb1Cre+ mice were intraperitoneally injected with 7.5 million of CD4+ T cell from C57BL/6 mice (control) or from bm12 mice. a,ELISA of anti-dsDNA in serum from immunized mice at day 14. b, Representative immunofluorescent images showing IgG deposits in kidney by staining with Cy3-labeled anti-mouse IgG. Scale bar, 50 μm. Data are means ± SEM. Student’s t tests were used (NS, not significant, *p < 0.05, **p < 0.01, ***p < 0.001). Data are pooled from 2 experiments.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed immunoblots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed immunoblots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 6

Unprocessed immunoblots.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed immunoblots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed immunoblots.

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. 5

Unprocessed immunoblots.

Source Data Extended Data Fig. 7

Unprocessed immunoblots.

Source Data Extended Data Fig. 8

Unprocessed immunoblots.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Springer Nature or its licensor 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

Yin, Y., Yang, X., Wu, S. et al. Jmjd1c demethylates STAT3 to restrain plasma cell differentiation and rheumatoid arthritis. Nat Immunol 23, 1342–1354 (2022). https://doi.org/10.1038/s41590-022-01287-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01287-y

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