Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2018 Aug 31;128(9):3779-3793.
doi: 10.1172/JCI98786. Epub 2018 Jul 30.

SUMO-defective c-Maf preferentially transactivates Il21 to exacerbate autoimmune diabetes

Affiliations

SUMO-defective c-Maf preferentially transactivates Il21 to exacerbate autoimmune diabetes

Chao-Yuan Hsu et al. J Clin Invest. .

Abstract

SUMOylation is involved in the development of several inflammatory diseases, but the physiological significance of SUMO-modulated c-Maf in autoimmune diabetes is not completely understood. Here, we report that an age-dependent attenuation of c-Maf SUMOylation in CD4+ T cells is positively correlated with the IL-21-mediated diabetogenesis in NOD mice. Using 2 strains of T cell-specific transgenic NOD mice overexpressing wild-type c-Maf (Tg-WTc) or SUMOylation site-mutated c-Maf (Tg-KRc), we demonstrated that Tg-KRc mice developed diabetes more rapidly than Tg-WTc mice in a CD4+ T cell-autonomous manner. Moreover, SUMO-defective c-Maf preferentially transactivated Il21 to promote the development of CD4+ T cells with an extrafollicular helper T cell phenotype and expand the numbers of granzyme B-producing effector/memory CD8+ T cells. Furthermore, SUMO-defective c-Maf selectively inhibited recruitment of Daxx/HDAC2 to the Il21 promoter and enhanced histone acetylation mediated by CREB-binding protein (CBP) and p300. Using pharmacological interference with CBP/p300, we illustrated that CBP30 treatment ameliorated c-Maf-mediated/IL-21-based diabetogenesis. Taken together, our results show that the SUMOylation status of c-Maf has a stronger regulatory effect on IL-21 than the level of c-Maf expression, through an epigenetic mechanism. These findings provide new insights into how SUMOylation modulates the pathogenesis of autoimmune diabetes in a T cell-restricted manner and on the basis of a single transcription factor.

Keywords: Autoimmunity; Cytokines; Epigenetics; Immunology; T cells.

PubMed Disclaimer

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

Figures

Figure 1
Figure 1. c-Maf SUMOylation in CD4+ T cells is inversely correlated with the severity of insulitis and IL-21 production in NOD mice.
(A) The severity of insulitis was classified and scored on 100 islets from 10 NOD mice per group. Immunoprecipitation analysis of c-Maf SUMOylation in 6- to 8-week-old and 12- to 14-week-old NOD CD4+ cells cultured with anti-CD3 and anti-CD28 for 36 hours. (B and C) Expressions of Sae1, Sae2, Ubc9, and Pias1 mRNA (B) or Il4, Il10, and Il21 mRNA (C) in CD4+ cells cultured for 36 hours as described in A. (D) ELISA of indicated cytokines in supernatants of CD4+ T cells cultured as described in A for 48 hours. (E) ChIP analysis of the interaction of c-Maf with the Il21 promoter (Il21p) in CD4+ cells cultured for 36 hours as described in A. (F) Immunoprecipitation analysis of c-Maf SUMOylation in 6- to 8-week-old NOD CD4+ cells cultured for 36 hours with anti-CD3 and anti-CD28 in the presence of anacardic acid (3 μM) or its solvent (DMSO), which were added after 18 hours of culture. (G) Expressions of indicated cytokine mRNA in CD4+ T cells cultured for 36 hours as described in F. (H) ELISA of indicated cytokines in supernatants of CD4+ cells cultured as described in F for 48 hours. (I) ChIP analysis of the interaction of c-Maf with the Il21 promoter in CD4+ cells cultured for 36 hours as described in F. For E and I, isotype-matched IgG was used as a control. See complete unedited blots in the supplemental material. Data represent the mean ± SEM; n = 5 mice (A and F), n = 3 mice (BE and GI) per group; 3 independent experiments. *P < 0.05; **P < 0.01; 2-tailed Student’s t test (AD and FH) or 1-way ANOVA with Tukey’s post-test (E and I).
Figure 2
Figure 2. Accelerated kinetics of autoimmune diabetes in SUMO-defective c-Maf–transgenic NOD mice.
(A) Schematic diagram of distal Lck promoter–driven wild-type c-Maf or K33R c-Maf transgenes. (B) PCR detection of transgenic c-Maf in genomic DNA extracted from control, Tg-WTc, and Tg-KRc mice. Il23 p19 was used as an internal control. (C) Western blot analysis of c-Maf in control, Tg-WTc, and Tg-KRc CD4+ T cells. (D and E) Immunoprecipitation analysis of c-Maf SUMOylation in control, Tg-WTc, and Tg-KRc CD4+ T cells cultured with anti-CD3 and anti-CD28 for 36 hours. (F) Diabetes incidence in Tg-WTc NOD mice and their littermate controls. (G) Diabetes incidence in Tg-KRc NOD mice and their littermate controls. (H) Diabetes incidence in NOD-SCID recipients of splenocytes from 12- to 14-week-old control, Tg-WTc, or Tg-KRc NOD mice. (I and J) CD25CD4+ plus CD8+ T cells from 12- to 14-week-old indicated NOD mice were transferred into NOD.Rag1–/– mice; all groups were treated simultaneously in 2 independent experiments. The control CD4/control CD8 (white circles) group is presented in both I and J. Diabetic incidences among groups that received control CD8+ T cells plus different CD4+ T cells (I) and diabetic incidences in among groups that received control CD4+ T cells plus different CD8+ T cells (J) were compared with each other by a log-rank test. See complete unedited blots in the supplemental material. For E, data represent the mean ± SEM; n = 3 mice (B and C) or n = 5 mice (D and E) per group; 3 independent experiments (BE) or 2 independent experiments (H). *P < 0.05; **P < 0.01; 1-way ANOVA with Tukey’s post-test (E) or log-rank test (FJ).
Figure 3
Figure 3. Overexpression of SUMO-defective c-Maf preferentially augments IL-21–producing CD4+ T cells by enhancing its recruitment to the Il21p.
(A and B) Frequencies of IFN-γ+, IL-4+, IL-10+, IL-17+, and IL-21+ cells (A) and Tg/control ratio of IFN-γ+, IL-4+, and IL-21+ cells (B) in splenic CD4+ T cells from 12- to 14-week-old control, Tg-WTc, and Tg-KRc NOD mice were analyzed by flow cytometry. (C and D) Frequencies (C) and Tg/control ratio (D) of IFN-γ+, IL-4+, and IL-21+ cells in pancreas-infiltrating CD4+ T cells from 12- to 14-week-old control, Tg-WTc, and Tg-KRc NOD mice were analyzed by flow cytometry. (E) Expressions of Il4, Il10, and Il21 mRNA in naive control, Tg-WTc, and Tg-KRc NOD CD4+ T cells cultured with anti-CD3 and anti-CD28 for 36 hours. (F) Intracellular staining for IL-4, IL-10, and IL-21 in naive CD4+ T cells cultured as described in E for 48 hours. Numbers in outlined areas indicate the percentages of the gated populations. (G) ELISA of IL-4, IL-10, and IL-21 in supernatants of naive CD4+ T cells cultured as described in E for 48 hours. (H and I) ChIP analysis of the interaction of c-Maf with the Il21p in naive CD4+ T cells cultured for 36 hours as described in E. Isotype-matched IgG was used as a control. Data represent the mean ± SEM; n = 6 mice (A and B), n = 10 mice (C and D), n = 3 mice (EG), or n = 5 mice (H and I) per group; 3–4 independent experiments. *P < 0.05; **P < 0.01; 1-way ANOVA with Tukey’s post-test (A, C, E, and GI) or 2-tailed Student’s t test (B and D).
Figure 4
Figure 4. SUMO-defective c-Maf promotes the differentiation of CD4+ T cells with an extrafollicular helper T cell phenotype.
(A) Flow cytometry analysis of the expression of ICOS, PD-1, and CXCR5 in splenic CD4+ T cells from 12- to 14-week-old control, Tg-WTc, and Tg-KRc NOD mice. Summary of the frequencies of ICOShiCD4+, PD-1hiCD4+, and CXCR5+CD4+ T cells. (B) Flow cytometry analysis of the expression of ICOS and PD-1 in splenic CD4+ T cells as described in A. Numbers adjacent to outlined areas indicate the percentages of ICOShiPD-1hi cells (left panel). Right panel: Summary of the frequencies of ICOShiPD-1hiCD4+ T cells. (C) Flow cytometry analysis of ICOS or PD-1 expression in ICOSloPD-1loCD4+ and ICOShiPD-1hiCD4+ T cells from 12- to 14-week-old control, Tg-WTc, and Tg-KRc NOD mice. Summary of the geometric mean fluorescence intensity (gMFI) of ICOS or PD-1 in ICOSloPD-1loCD4+ or ICOShiPD-1hiCD4+ T cells. (D) Flow cytometry analysis of the expression of ICOS, PD-1, and CXCR5 in splenic CD4+ T cells as described in A. Summary of the frequencies of CXCR5ICOShiPD-1hiCD4+ and CXCR5+ICOShiPD-1hiCD4+ T cells. (E and F) Expression of Il21 mRNA in CXCR5ICOSloPD-1loCD4+, CXCR5ICOShiPD-1hiCD4+, and CXCR5+ICOShiPD-1hiCD4+ T cells from control, Tg-WTc, and Tg-KRc NOD mice. Data represent the mean ± SEM; n = 6 mice (AD) or n = 3 mice (E and F) per group; 3–4 independent experiments. *P < 0.05; **P < 0.01; 1-way ANOVA with Tukey’s post-test.
Figure 5
Figure 5. SUMO-defective c-Maf expands effector/memory CD8+ T cells and enhances their granzyme B production and diabetogenic activity in an IL-21–dependent manner.
(A) Flow cytometry analysis of CD69, KLRG1, and CD44 in splenic CD8+ T cells from 12- to 14-week-old control, Tg-WTc, and Tg-KRc NOD mice. Summary of the frequencies of CD69+CD8+, KLRG1+CD8+, and CD44hiCD8+ T cells. (B) Expression of Ifng, Gzmb, and Prf1 mRNA in splenic CD8+ T cells from 12- to 14-week-old control, Tg-WTc, and Tg-KRc NOD mice. (C) Expression of Gzmb mRNA in CD44loCD8+ and CD44hiCD8+ T cells from 12- to 14-week-old control, Tg-WTc, and Tg-KRc NOD mice. (D) Intracellular staining for granzyme B in control CD8+ T cells cocultured with control, Tg-WTc, and Tg-KRc CD4+ cells in Transwell chambers for 48 hours with anti-CD3 and anti-CD28 in the presence of control.Fc and IL-21R.Fc (2 μg/ml). Numbers adjacent to outlined areas indicate the percentages of granzyme B+ cells. Summary of the frequency of granzyme B+CD8+ T cells. (E) Diabetes incidence in Tg-KRc.Il21r+/+ and Tg-KRc.Il21r–/– NOD mice. (F) Diabetes incidence in NOD.Rag1–/– recipients injected with Tg-WTc and Tg-KRc CD25CD4+ T cells plus control CD8+ T cells on day 0, and then injected with control.Fc or IL-21R.Fc (10 μg) every 2 days from day 1 to day 13. (G) On day 14, quantitative reverse transcription PCR (RT-qPCR) analysis of Cd44 or Gzmb mRNA expression in CD8+ T cells from NOD.Rag1−/− recipients reconstituted as described in F. Data represent the mean ± SEM; n = 6 mice (A), n = 4 mice (BD), n = 3 mice (G) per group; 3 independent experiments (AD) or 2 independent experiments (F and G). *P < 0.05; **P < 0.01; 1-way ANOVA with Tukey’s post-test (A, B, D, and G), 2-tailed Student’s t test (C), or log-rank test (E and F).
Figure 6
Figure 6. SUMO-defective c-Maf prevents Daxx/HDAC2 recruitment to the Il21p and enhances CBP/p300-mediated histone acetylation.
(AC) ChIP analysis of the interaction of Daxx (A), HDAC1 (B), and HDAC2 (C) with the c-Maf–binding sites of the Il4p and the Il21p in naive control, Tg-WTc, and Tg-KRc CD4+ T cells cultured with anti-CD3 and anti-CD28 for 36 hours. (D and E) ChIP analysis of the abundance of H3ac (D) and H4ac (E) in the c-Maf–binding site of the Il4p or the Il21p in naive CD4+ T cells cultured for 36 hours as in AC. (F and G) ChIP analysis of the interaction of CBP (F) and p300 (G) with the c-Maf–binding site of the Il4p or the Il21p in naive CD4+ T cells cultured for 36 hours as in AC. Isotype-matched IgG was used as a control. Data represent the mean ± SEM; n = 3–5 mice per group; 3 independent experiments. *P < 0.05; **P < 0.01; 1-way ANOVA with Tukey’s post-test.
Figure 7
Figure 7. A CBP/p300 inhibitor attenuates SUMO-defective c-Maf–mediated transactivation of Il21 and ameliorates autoimmune diabetes in NOD mice.
(A) Expression of Il21 mRNA in naive control, Tg-WTc, and Tg-KRc CD4+ T cells cultured for 36 hours with anti-CD3 and anti-CD28 in the presence of CBP30 (2 μM) or its solvent (DMF), which were added after 18 hours of culture. (BD) ChIP analysis of the interaction of c-Maf (B), CBP (C), and p300 (D) with the c-Maf–binding site in the Il21p in naive CD4+ T cells cultured for 36 hours as in A. (E and F) ChIP analysis of the abundance of H3ac (E) and H4ac (F) in the c-Maf–binding site of the Il21p in naive CD4+ T cells cultured for 36 hours as in A. Isotype-matched IgG was used as a control. (G) Diabetic incidence in NOD.Rag1–/– recipients injected with effector Tg-WTc and Tg-KRc CD4+ T cells (CD4+CD25) plus control CD8+ T cells on day 0, and then injected with 2 mg/kg CBP30 or its solvent (DMF) every 2 days from day 1 to day 13. (H) On day 14, RT-qPCR analysis of Il21 mRNA expression in CD4+ T cells and Gzmb mRNA expression in CD8+ T cells from NOD.Rag1–/– recipients reconstituted as in G. Data represent the mean ± SEM; n = 3 mice (A and H), n = 5 mice (BF), n = 5–6 mice (G) per group; 3 independent experiments (AF) or 2 independent experiments (G and H). *P < 0.05; **P < 0.01; 2-tailed Student’s t test (AF), log-rank test (G), or 1-way ANOVA with Tukey’s post-test (H).
Figure 8
Figure 8. Schematic diagrams illustrating the critical role of SUMOylation in the regulation of c-Maf–mediated IL-21 production.

References

    1. Johnson ES. Protein modification by SUMO. Annu Rev Biochem. 2004;73:355–382. doi: 10.1146/annurev.biochem.73.011303.074118. - DOI - PubMed
    1. Liu B, Tahk S, Yee KM, Fan G, Shuai K. The ligase PIAS1 restricts natural regulatory T cell differentiation by epigenetic repression. Science. 2010;330(6003):521–525. doi: 10.1126/science.1193787. - DOI - PMC - PubMed
    1. Decque A, et al. Sumoylation coordinates the repression of inflammatory and anti-viral gene-expression programs during innate sensing. Nat Immunol. 2016;17(2):140–149. - PubMed
    1. Ding X, et al. Protein SUMOylation is required for regulatory T cell expansion and function. Cell Rep. 2016;16(4):1055–1066. doi: 10.1016/j.celrep.2016.06.056. - DOI - PubMed
    1. Geiss-Friedlander R, Melchior F. Concepts in sumoylation: a decade on. Nat Rev Mol Cell Biol. 2007;8(12):947–956. doi: 10.1038/nrm2293. - DOI - PubMed

Publication types

MeSH terms