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:

Targeting JMJD1C to selectively disrupt tumor Treg cell fitness enhances antitumor immunity

Abstract

Regulatory T (Treg) cells are critical for immune tolerance but also form a barrier to antitumor immunity. As therapeutic strategies involving Treg cell depletion are limited by concurrent autoimmune disorders, identification of intratumoral Treg cell-specific regulatory mechanisms is needed for selective targeting. Epigenetic modulators can be targeted with small compounds, but intratumoral Treg cell-specific epigenetic regulators have been unexplored. Here, we show that JMJD1C, a histone demethylase upregulated by cytokines in the tumor microenvironment, is essential for tumor Treg cell fitness but dispensable for systemic immune homeostasis. JMJD1C deletion enhanced AKT signals in a manner dependent on histone H3 lysine 9 dimethylation (H3K9me2) demethylase and STAT3 signals independently of H3K9me2 demethylase, leading to robust interferon-γ production and tumor Treg cell fragility. We have also developed an oral JMJD1C inhibitor that suppresses tumor growth by targeting intratumoral Treg cells. Overall, this study identifies JMJD1C as an epigenetic hub that can integrate signals to establish tumor Treg cell fitness, and we present a specific JMJD1C inhibitor that can target tumor Treg cells without affecting systemic immune homeostasis.

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

Fig. 1: Expression of Jmjd1c is upregulated in tumor Treg cells by a combination of cytokines.
Fig. 2: Treg cell Jmjd1c deficiency reduces tumor Treg cell frequency and inhibits tumor growth.
Fig. 3: JMJD1C suppresses AKT signaling by promoting NRP1 and PD1 expression to prevent IFNγ production.
Fig. 4: JMJD1C demethylates and inhibits STAT3 in tumor Treg cells to prevent IFNγ production.
Fig. 5: IFNγ deletion rescues tumor Treg cell fragility in the absence of JMJD1C.
Fig. 6: JMJD1C inhibitor suppresses tumor growth by targeting tumor Treg cells.

Similar content being viewed by others

Data availability

scRNA-seq data have been deposited in the GEO database under accession code GSE223930. ATAC-seq and CUT&RUN-seq data have been deposited in GEO under accession code GSE224084. Publicly available datasets accessed for use in this manuscript include GSE139325, GSE108989, GSE98638 and GSE99254. Source data are provided with this paper.

Code availability

The codes for scRNA-seq, ATAC-seq and CUT&RUN data analyses reported in this study have been deposited at GitHub (https://github.com/YuliangWang316/JMJD1C_Treg). Any additional information required to re-analyze the data is available from the corresponding authors upon request.

References

  1. Sakaguchi, S. et al. Regulatory T cells and human disease. Annu. Rev. Immunol. 38, 541–566 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Savage, P. A., Klawon, D. E. J. & Miller, C. H. Regulatory T cell development. Annu. Rev. Immunol. 38, 421–453 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Tanaka, A. & Sakaguchi, S. Targeting Treg cells in cancer immunotherapy. Eur. J. Immunol. 49, 1140–1146 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Plitas, G. & Rudensky, A. Y. Regulatory T cells in cancer. Annu. Rev. Cancer Biol. 4, 459–477 (2020).

    Article  Google Scholar 

  5. Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 24, 541–550 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Togashi, Y., Shitara, K. & Nishikawa, H. Regulatory T cells in cancer immunosuppression-implications for anticancer therapy. Nat. Rev. Clin. Oncol. 16, 356–371 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Sharma, P. & Allison, J. P. The future of immune checkpoint therapy. Science 348, 56–61 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  8. Tanaka, A. & Sakaguchi, S. Regulatory T cells in cancer immunotherapy. Cell Res. 27, 109–118 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Lim, S. A. et al. Lipid signalling enforces functional specialization of Treg cells in tumours. Nature 591, 306–311 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, H. et al. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat. Immunol. 21, 298–308 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Chuckran, C. A., Liu, C., Bruno, T. C., Workman, C. J. & Vignali, D. A. Neuropilin-1: a checkpoint target with unique implications for cancer immunology and immunotherapy. J. Immunother. Cancer 8, e000967 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Delgoffe, G. M. et al. Stability and function of regulatory T cells is maintained by a neuropilin-1–semaphorin-4a axis. Nature 501, 252–256 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Overacre-Delgoffe, A. E. et al. Interferon-γ drives Treg fragility to promote anti-tumor immunity. Cell 169, 1130–1141.e1111 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hatzioannou, A. et al. An intrinsic role of IL-33 in Treg cell-mediated tumor immunoevasion. Nat. Immunol. 21, 75–85 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Kim, M. J. et al. Deletion of PD-1 destabilizes the lineage identity and metabolic fitness of tumor-infiltrating regulatory T cells. Nat. Immunol. 24, 148–161 (2023).

    Article  CAS  PubMed  Google Scholar 

  16. Kidani, Y. et al. CCR8-targeted specific depletion of clonally expanded Treg cells in tumor tissues evokes potent tumor immunity with long-lasting memory. Proc. Natl Acad. Sci. USA 119, e2114282119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. De Simone, M. et al. Transcriptional landscape of human tissue lymphocytes unveils uniqueness of tumor-infiltrating T regulatory cells. Immunity 45, 1135–1147 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Ahuja, N., Sharma, A. R. & Baylin, S. B. Epigenetic therapeutics: a new weapon in the war against cancer. Annu. Rev. Med. 67, 73–89 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Feng, Y. et al. Control of the inheritance of regulatory T cell identity by a cis element in the Foxp3 locus. Cell 158, 749–763 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Placek, K. et al. MLL4 prepares the enhancer landscape for Foxp3 induction via chromatin looping. Nat. Immunol. 18, 1035–1045 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. DuPage, M. et al. The chromatin-modifying enzyme Ezh2 is critical for the maintenance of regulatory T cell identity after activation. Immunity 42, 227–238 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Singh Nanda, J., Kumar, R. & Raghava, G. P. S. dbEM: a database of epigenetic modifiers curated from cancerous and normal genomes. Sci. Rep. 6, 19340 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  24. Guo, X. et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 24, 978–985 (2018).

    Article  CAS  PubMed  Google Scholar 

  25. Zheng, C. et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell 169, 1342–1356.e1316 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Montauti, E. et al. A deubiquitination module essential for Treg fitness in the tumor microenvironment. Sci. Adv. 8, eabo4116 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Akimova, T. et al. Human lung tumor FOXP3+ Tregs upregulate four ‘Treg-locking’ transcription factors. JCI Insight 2, e94075 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Kartikasari, A. E. R., Huertas, C. S., Mitchell, A. & Plebanski, M. Tumor-induced inflammatory cytokines and the emerging diagnostic devices for cancer detection and prognosis. Front. Oncol. 11, 692142 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Fisher, D. T., Appenheimer, M. M. & Evans, S. S. The two faces of IL-6 in the tumor microenvironment. Semin. Immunol. 26, 38–47 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rébé, C. & Ghiringhelli, F. Interleukin-1β and cancer. Cancers 12, 331–344 (2020).

    Article  Google Scholar 

  31. Kesanakurti, D., Chetty, C., Rajasekhar Maddirela, D., Gujrati, M. & Rao, J. S. Essential role of cooperative NF-κB and Stat3 recruitment to ICAM-1 intronic consensus elements in the regulation of radiation-induced invasion and migration in glioma. Oncogene 32, 5144–5155 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Lee, H. et al. Persistently activated Stat3 maintains constitutive NF-κB activity in tumors. Cancer Cell 15, 283–293 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Choi, Y., Kim, J. K. & Yoo, J. Y. NFκB and STAT3 synergistically activate the expression of FAT10, a gene counteracting the tumor suppressor p53. Mol. Oncol. 8, 642–655 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zheng, L. et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 374, abe6474 (2021).

    Article  PubMed  Google Scholar 

  35. Yin, Y. et al. Jmjd1c demethylates STAT3 to restrain plasma cell differentiation and rheumatoid arthritis. Nat. Immunol. 23, 1342–1354 (2022).

    Article  CAS  PubMed  Google Scholar 

  36. Komatsu, N. et al. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat. Med. 20, 62–68 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Laurence, A. et al. STAT3 transcription factor promotes instability of nTreg cells and limits generation of iTreg cells during acute murine graft-versus-host disease. Immunity 37, 209–222 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  39. 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 

  40. Gocher, A. M., Workman, C. J. & Vignali, D. A. A. Interferon-γ: teammate or opponent in the tumour microenvironment? Nat. Rev. Immunol. 22, 158–172 (2022).

    Article  CAS  PubMed  Google Scholar 

  41. Boehm, U., Klamp, T., Groot, M. & Howard, J. C. Cellular responses to interferon-γ. Annu. Rev. Immunol. 15, 749–795 (1997).

    Article  CAS  PubMed  Google Scholar 

  42. Zagorulya, M. et al. Tissue-specific abundance of interferon-gamma drives regulatory T cells to restrain DC1-mediated priming of cytotoxic T cells against lung cancer. Immunity 56, 386–405.e10 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ni, X. et al. YAP is essential for Treg-mediated suppression of antitumor immunity. Cancer Discov. 8, 1026–1043 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhang, L. et al. Histone methyltransferase Nsd2 ensures maternal-fetal immune tolerance by promoting regulatory T-cell recruitment. Cell. Mol. Immunol. 19, 634–643 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Batzner, S. et al. E(3)-equivariant graph neural networks for data-efficient and accurate interatomic potentials. Nat. Commun. 13, 2453 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Shen, C. et al. The impact of cross-docked poses on performance of machine learning classifier for protein-ligand binding pose prediction. J. Cheminform. 13, 81 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Imrie, F., Bradley, A. R. & Deane, C. M. Generating property-matched decoy molecules using deep learning. Bioinformatics 37, 2134–2141 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Mysinger, M. M., Carchia, M., Irwin, J. J. & Shoichet, B. K. Directory of useful decoys, enhanced (DUD-E): better ligands and decoys for better benchmarking. J. Med. Chem. 55, 6582–6594 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bauer, M. R., Ibrahim, T. M., Vogel, S. M. & Boeckler, F. M. Evaluation and optimization of virtual screening workflows with DEKOIS 2.0-a public library of challenging docking benchmark sets. J. Chem. Inf. Model. 53, 1447–1462 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Z. Qin (Institute of Biophysics, Chinese Academy of Sciences) for providing MCA205 cells. This study was supported by the National Natural Science Foundation of China (grant numbers 32330036 to X.W., T222502 to M.Z., 81825018 to J.Q., 82301970 to X. Long, 82130085 to J.Q., 82101827 to Jingjing Chen, 82273855 to M.Z., 82204278 to X. Li and 31970828 to X.W.), National Key Research and Development Program of China (2022YFC3400504 to M.Z.), Youth Innovation Promotion Association CAS (2023296 to S. Zhang), Lingang Laboratory (LG202102-01-02 to M.Z. and LG-QS-202204-01 to S. Zhang), China Postdoctoral Science Foundation (2022M721675 to X. Long, 2020M681665 to Jingjing Chen and 2022M720153 to X. Li), Postgraduate Research & Practice Innovation Program of Jiangsu Province (KYCX22_1784 to Z.Z. and JX10113833 to S.W.), Postdoc Research Program of Taizhou School of Clinical Medicine (TZBSHKY202203 to X. Long), Major Program of Wuxi Medical Center, Nanjing Medical University (WMCM202310 to X.W.), Jiangsu Provincial Key Research Development Program of China (BE2022770 to Y.C.) and Jiangsu Outstanding Young Investigator Program (BK20200030 to X.W.).

Author information

Authors and Affiliations

Authors

Contributions

X.W., J.Q. and M.Z. conceived and directed the study. X. Long, Y.W., Jingjing Chen, Y.L., Y.C. and B.L. performed most of the animal and cellular biology experiments. S. Zhang, H.H., X. Li, C.S., R.Y., D.C., G.C. and D.W. designed and performed the experiments regarding the small compound. R.C. performed the pharmacokinetic study of the inhibitor. Y.W. performed the bioinformatic analysis. H.Z., S. Zhai, Z.Z., S.W., M.L., J.Z. and Junhong Chen helped with mouse care and some in vitro experiments. X.W., J.Q. and M.Z. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Mingyue Zheng, Jun Qin or Xiaoming Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Axel Kallies and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: N. Bernard, in collaboration with the Nature Immunology team.

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 Slingshot analysis based pseudotime analysis.

Slingshot analysis based pseudotime analysis was derived for the three populations of Treg from Fig. 1g.

Extended Data Fig. 2 JMJD1C is dispensable for systemic Treg development under steady state.

a, Splenic Treg cells were sorted out from Jmjd1cTreg WT and Jmjd1cTreg KO, treated with TNF plus IL-6 for 3 days, and subject to western blot analysis with anti-JMJD1C. b, Comparison of Treg cell cellularity in spleen and lymph nodes between Jmjd1cTreg WT (n = 7) and Jmjd1cTreg KO (n = 6) mice. pLN, peripheral lymphoid nodes. c, Representative flow cytometric plots of splenic CD4+ T cell subsets (up) or CD8+ T cell subsets(down) : naïve T (CD44 CD62L+) and T effector memory (CD44+CD62L) cells from Jmjd1cTreg WT (n = 7) and Jmjd1cTreg KO (n = 6) mice. d, Representative images of hematoxylin and eosin staining for the indicated tissues from Jmjd1cTreg WT or Jmjd1cTreg KO mice at the age of 6-month-old. Scale bars: spleen 500 µm, colon 500 µm, lung 200 µm, liver 100 µm, kidney 50 µm. e, Frequency of Treg cells in different tissues as indicated from 6-month-old mice. Data represent two (a) or three (d) independent experiments. Data were pooled from 2 independent experiments (b, c, e). Data in (b, c, e) are shown in means ± SD. Two-tailed unpaired Student’s t-test (b, c, e).

Source data

Extended Data Fig. 3 Characterization of tumor Treg expansion, apoptosis and suppressive function.

a, Gating strategy for tumor Treg. b,c, Representative flow cytometric plots of Ki67 (b) and active caspase3 (c) levels in splenic Treg cells or intratumoral Treg cells from MCA205-tumor-bearing Jmjd1cTreg WT (n = 6) and Jmjd1cTreg KO (n = 7) mice. d, e, Ex vivo suppression of CellTrace Violet (CTV)-labeled WT naïve CD8+ T cell proliferation by Jmjd1cTreg WT and Jmjd1cTreg KO Treg cells sorted from tumors (d) or spleens (e) with the annotated co-culture ratios. Teff, CD8+ T effector cells. n = 3 biological independent samples. Data represent three independent experiments (b-e). Data in (b-e) are shown in means ± SD. Two-tailed unpaired Student’s t-test (b-e).

Source data

Extended Data Fig. 4 scRNA-seq analysis of tumor Treg cells from MCA205-tumor-bearing mice.

a, Unsupervised clustering of tumor-infiltrating Treg cells from MCA205-tumor-bearing Jmjd1cTreg WT (4 mice) and Jmjd1cTreg KO (7 mice) mice. b, Dot plot showing the expression of marker genes for each cluster. c, Tumor Treg cells from MCA205-tumor-bearing Jmjd1cTreg WT (n = 6 mice) and Jmjd1cTreg KO mice (n = 7 mice) were stained for 4-1BB expression by flow cytometry. d, Tumor-infiltrating Treg cells cells were sorted from MCA205-tumor-bearing Jmjd1cTreg WT and Jmjd1cTreg KO mice, the protein levels of H3K9me2 and H3K9me1 were assessed by western blot. e, Volcano plot showing the genes differentially expressed in Jmjd1c WT and KO tumor Treg cells from the scRNA-seq data. P values were computed with two-sided Wilcoxon rank-sum test in Seurat and adjusted with Bonferroni method. f, Gene Set Enrichment Analysis (GSEA) showing the significantly enriched signatures among all the 50 hallmark gene sets in Jmjd1c WT versus Jmjd1c KO tumor Treg cells. P values were computed with empirical phenotype-based permutation tests (GSEA), and adjusted for multiple comparisons using the Benjamini–Hochberg method (FDR). Data represent two independent experiments (c, d). Data are shown in means ± SD (c). Two-tailed unpaired Student’s t-test (c).

Source data

Extended Data Fig. 5 Cytokine production in tumor effector T cells from WT and Stat3CATreg OE mice.

a, Representative flow cytometric plots of IFNγ and TNF production in Foxp3CD4+ and CD8+ tumor-infiltrating T cells (left). Percentage of cytokine-producing T cells in dLN and tumor were plotted on the right. n = 5 mice. b, Representative flow cytometric plots of IL-17A production in Foxp3-CD4+ tumor-infiltrating T cells (left). Percentage of IL-17A-producing T cells were plotted on the right. n = 5 mice. Data represent two independent experiments. Data in (a, b) are shown in means ± SD. Two-tailed unpaired Student’s t-test (a-b).

Source data

Extended Data Fig. 6 193D7 treatment suppresses tumor growth.

C57BL6/J mice were subcutaneously inoculated with B16-F10, LLC or Hepa1-6 tumor cells. BALB/C mice were subcutaneously inoculated with CT26 tumor cells. When tumor grew to measurable size (~100 mm3), the tumor bearing mice were intraperitoneally treated with DMSO control or 193D7 at 25 mg/kg once a day, tumor growth was monitored. n = 6 mice. Data represent two independent experiments. Data are shown in means ± SD. Two-way ANOVA was used.

Source data

Extended Data Fig. 7 193D7 treatment shows no evidence of toxicity in mice.

C57BL6/J mice were intraperitoneally treated with DMSO control or 193D7 at 25mg/kg once a day for 14 days. n = 6 mice per group. a, Body weight was monitored over time. b, Quantification of alanine aminotransferase (ALT), alkaline phosphatase (ALP) in the serum at day 14. c, Representative images of hematoxylin and eosin staining for the indicated tissues from DMSO control or 193D7-treated mice at day 14. Scale bars: colon 500 µm, lung 200µm, liver 100 µm, kidney 50 µm. Data represent two independent experiments (a-c). Data are shown in means ± SD (a, b). Two-way ANOVA (a); two-tailed unpaired Student’s t-test (b).

Source data

Extended Data Fig. 8 193D7 treatment increased tumor effector T cell number and cytokine production by in wildtype mice.

C57BL6/J mice bearing MCA205 tumor were orally treated with DMSO control or 193D7 at 25 mg/kg once a day for 12 days as in Fig. 6j. DMSO, n = 7 mice; 193D7, n = 8 mice. a, Cell number of CD4+ and CD8+ T cells per gram tumor in MCA205-tumor-bearing mice. b, Percentage of cytokine-producing T cells in dLNs and tumors. Data represent two independent experiments. Data are shown in means ± SD. Two-tailed unpaired Student’s t-test (a, b).

Source data

Extended Data Fig. 9 193D7 treatment did not alter tumor effector T cell number and cytokine production in Jmjd1cTreg KO mice.

Jmjd1cTreg KO mice bearing MCA205 tumor were orally treated with DMSO control or 193D7 at 25mg/kg once a day for 12 days as in Fig. 6p. n = 5 mice. a, Cell number of CD4+ and CD8+ T cells per gram tumor in MCA205-tumor-bearing mice. b, Percentage of cytokine-producing T cells in tumors. Data represent two independent experiments. Data are shown in means ± SD (a, b). Two-tailed unpaired Student’s t-test (a, b).

Source data

Extended Data Fig. 10 Analysis of the effect of 193D7 treatment on tumor Treg cells in mixed bone marrow chimeras.

a, The experimental design of mixed bone marrow chimeras. b, The Treg frequency in tumor was analyzed by flow cytometry in tumor-bearing mixed chimeras. n = 6 mice. c, Lymphocytes were isolated from tumor tissues, stimulated and stained for IFNγ in tumor Treg cells. n = 6 mice. Data represent two independent experiments. Plotting data in (b, c) are all shown in means ± SD. Two-tailed unpaired Student’s t-test (b, c).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–7.

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Table 1: list of genes with alterations in H3K9me2 signal and mRNA expression. Supplementary Table 2: top 100 candidate JMJD1C inhibitors.

Supplementary Data 1

Source data for Supplementary Fig. 1.

Supplementary Data 2

Source data for Supplementary Fig. 2.

Supplementary Data 3

Source data for Supplementary Fig. 3.

Supplementary Data 4

Source data for Supplementary Fig. 4.

Supplementary Data 5

Source data for Supplementary Fig. 5.

Supplementary Data 6

Source data for Supplementary Fig. 6.

Supplementary Data 7

Source data for Supplementary Fig. 7.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

Long, X., Zhang, S., Wang, Y. et al. Targeting JMJD1C to selectively disrupt tumor Treg cell fitness enhances antitumor immunity. Nat Immunol 25, 525–536 (2024). https://doi.org/10.1038/s41590-024-01746-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-024-01746-8

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer