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:

Resetting the epigenetic balance of Polycomb and COMPASS function at enhancers for cancer therapy

Abstract

The lysine methyltransferase KMT2C (also known as MLL3), a subunit of the COMPASS complex, implements monomethylation of Lys4 on histone H3 (H3K4) at gene enhancers. KMT2C (hereafter referred to as MLL3) frequently incurs point mutations across a range of human tumor types, but precisely how these lesions alter MLL3 function and contribute to oncogenesis is unclear. Here we report a cancer mutational hotspot in MLL3 within the region encoding its plant homeodomain (PHD) repeats and demonstrate that this domain mediates association of MLL3 with the histone H2A deubiquitinase and tumor suppressor BAP1. Cancer-associated mutations in the sequence encoding the MLL3 PHD repeats disrupt the interaction between MLL3 and BAP1 and correlate with poor patient survival. Cancer cells that had PHD-associated MLL3 mutations or lacked BAP1 showed reduced recruitment of MLL3 and the H3K27 demethylase KDM6A (also known as UTX) to gene enhancers. As a result, inhibition of the H3K27 methyltransferase activity of the Polycomb repressive complex 2 (PRC2) in tumor cells harboring BAP1 or MLL3 mutations restored normal gene expression patterns and impaired cell proliferation in vivo. This study provides mechanistic insight into the oncogenic effects of PHD-associated mutations in MLL3 and suggests that restoration of a balanced state of Polycomb–COMPASS activity may have therapeutic efficacy in tumors that bear mutations in the genes encoding these epigenetic factors.

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: MLL3 COMPASS associates with the BAP1 complex.
Fig. 2: Cancer-associated MLL3PHD mutations disrupt MLL3–BAP1 binding and correlate with decreased patient survival.
Fig. 3: BAP1-dependent recruitment of MLL3 COMPASS to enhancers.
Fig. 4: MLL3 regulates tumor suppressor expression from BAP1-depdendent enhancers.
Fig. 5: BAP1 depletion leads to increased H3K27me3 due to loss of UTX-containing MLL3 COMPASS from chromatin.
Fig. 6: Mutations within MLL3PHD sensitize cells to PRC2 inhibition.

Similar content being viewed by others

References

  1. Morgan, M. A. & Shilatifard, A. Chromatin signatures of cancer. Genes Dev. 29, 238–249 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Plass, C. et al. Mutations in regulators of the epigenome and their connections to global chromatin patterns in cancer. Nat. Rev. Genet. 14, 765–780 (2013).

    Article  PubMed  CAS  Google Scholar 

  3. Hamamoto, R. et al. SMYD3 encodes a histone methyltransferase involved in the proliferation of cancer cells. Nat. Cell Biol. 6, 731–740 (2004).

    Article  PubMed  CAS  Google Scholar 

  4. Kotake, Y. et al. pRB family proteins are required for H3K27 trimethylation and Polycomb repression complexes binding to and silencing p16 INK4α tumor suppressor gene. Genes Dev. 21, 49–54 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Mazur, P. K. et al. SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer. Nature 510, 283–287 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Piunti, A. & Shilatifard, A. Epigenetic balance of gene expression by Polycomb and COMPASS families. Science 352, aad9780 (2016).

    Article  PubMed  CAS  Google Scholar 

  7. Ferguson, A. D. et al. Structural basis of substrate methylation and inhibition of SMYD2. Structure 19, 1262–1273 (2011).

    Article  PubMed  CAS  Google Scholar 

  8. Herz, H. M. et al. Histone H3 lysine-to-methionine mutants as a paradigm to study chromatin signaling. Science 345, 1065–1070 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Klaus, C. R. et al. DOT1L inhibitor EPZ-5676 displays synergistic anti-proliferative activity in combination with standard-of-care drugs and hypomethylating agents in MLL-rearranged leukemia cells. J. Pharmacol. Exp. Ther. 350, 646–656 (2014).

    Article  PubMed  CAS  Google Scholar 

  10. Kubicek, S. et al. Reversal of H3K9me2 by a small-molecule inhibitor for the G9a histone methyltransferase. Mol. Cell 25, 473–481 (2007).

    Article  PubMed  CAS  Google Scholar 

  11. Mohan, M., Herz, H. M. & Shilatifard, A. SnapShot: histone lysine methylase complexes. Cell 149, 498–498.e1 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    Article  PubMed  CAS  Google Scholar 

  13. Shilatifard, A. The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis. Annu. Rev. Biochem. 81, 65–95 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Miller, T. et al. COMPASS: a complex of proteins associated with a trithorax-related SET domain protein. Proc. Natl Acad. Sci. USA 98, 12902–12907 (2001).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Morgan, M. A. & Shilatifard, A. Drosophila SETs its sights on cancer: Trr/MLL3/MLL4 COMPASS-like complexes in development and disease. Mol. Cell. Biol. 33, 1698–1701 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Hu, D. & Shilatifard, A. Epigenetics of hematopoiesis and hematological malignancies. Genes Dev. 30, 2021–2041 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Yu, B. D., Hanson, R. D., Hess, J. L., Horning, S. E. & Korsmeyer, S. J. MLL, a mammalian trithorax-group gene, functions as a transcriptional maintenance factor in morphogenesis. Proc. Natl Acad. Sci. USA 95, 10632–10636 (1998).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Ortega-Molina, A. et al. The histone lysine methyltransferase KMT2D sustains a gene expression program that represses B cell lymphoma development. Nat. Med. 21, 1199–1208 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Zhang, J. et al. Disruption of KMT2D perturbs germinal center B cell development and promotes lymphomagenesis. Nat. Med. 21, 1190–1198 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Herz, H. M. et al. Enhancer-associated H3K4 monomethylation by trithorax-related, the Drosophila homolog of mammalian MLL3 and MLL4. Genes Dev. 26, 2604–2620 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Hu, D. et al. The MLL3 and MLL4 branches of the COMPASS family function as major histone H3K4 monomethylases at enhancers. Mol. Cell. Biol. 33, 4745–4754 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Fujimoto, A. et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat. Genet. 44, 760–764 (2012).

    Article  PubMed  CAS  Google Scholar 

  24. Ellis, M. J. et al. Whole-genome analysis informs breast cancer response to aromatase inhibition. Nature 486, 353–360 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Li, W. D. et al. Exome sequencing identifies an MLL3 gene germ line mutation in a pedigree of colorectal cancer and acute myeloid leukemia. Blood 121, 1478–1479 (2013).

    Article  PubMed  CAS  Google Scholar 

  26. Gui, Y. et al. Frequent mutations of chromatin-remodeling genes in transitional cell carcinoma of the bladder. Nat. Genet. 43, 875–878 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Chen, C. et al. MLL3 is a haploinsufficient 7q tumor suppressor in acute myeloid leukemia. Cancer Cell 25, 652–665 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Chaudhuri, A. R. et al. Replication fork stability confers chemoresistance in BRCA-deficient cells. Nature 535, 382–387 (2016).

    Article  PubMed Central  CAS  Google Scholar 

  29. Bailey, P. et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 531, 47–52 (2016).

    Article  PubMed  CAS  Google Scholar 

  30. Paoletti, A. C. et al. Quantitative proteomic analysis of distinct mammalian Mediator complexes using normalized spectral abundance factors. Proc. Natl Acad. Sci. USA 103, 18928–18933 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Ramón-Maiques, S. et al. The plant homeodomain finger of RAG2 recognizes histone H3 methylated at both lysine 4 and arginine 2. Proc. Natl Acad. Sci. USA 104, 18993–18998 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Carbone, M. et al. BAP1 and cancer. Nat. Rev. Cancer 13, 153–159 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Dey, A. et al. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science 337, 1541–1546 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Tripathi, S. et al. Meta- and orthogonal integration of influenza “OMICs” data defines a role for UBR4 in virus budding. Cell Host Microbe 18, 723–735 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Cieply, B., Farris, J., Denvir, J., Ford, H. L. & Frisch, S. M. Epithelial–mesenchymal transition and tumor suppression are controlled by a reciprocal feedback loop between ZEB1 and Grainyhead-like-2. Cancer Res. 73, 6299–6309 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Li, Y. et al. Downregulation of RBMS3 is associated with poor prognosis in esophageal squamous cell carcinoma. Cancer Res. 71, 6106–6115 (2011).

    Article  PubMed  Google Scholar 

  37. Nguyen, T. M. H. et al. Loss of ITM2A, a novel tumor suppressor of ovarian cancer through G2/M cell cycle arrest, is a poor prognostic factor of epithelial ovarian cancer. Gynecol. Oncol. 140, 545–553 (2016).

    Article  PubMed  CAS  Google Scholar 

  38. Cha, Y., Kim, D. K., Hyun, J., Kim, S. J. & Park, K. S. TCEA3 binds to TGF-β receptor 1 and induces SMAD-independent, JNK-dependent apoptosis in ovarian cancer cells. Cell. Signal. 25, 1245–1251 (2013).

    Article  PubMed  CAS  Google Scholar 

  39. Cui, T. et al. The p53 target gene desmocollin 3 acts as a novel tumor suppressor through inhibiting EGFR–ERK pathway in human lung cancer. Carcinogenesis 33, 2326–2333 (2012).

    Article  PubMed  CAS  Google Scholar 

  40. Qin, S., Zhang, Z., Li, J. & Zang, L. FRZB knockdown upregulates β-catenin activity and enhances cell aggressiveness in gastric cancer. Oncol. Rep. 31, 2351–2357 (2014).

    Article  PubMed  CAS  Google Scholar 

  41. Wang, S. et al. DACT2 is a functional tumor suppressor through inhibiting Wnt–β-catenin pathway and associated with poor survival in colon cancer. Oncogene 34, 2575–2585 (2015).

    Article  PubMed  CAS  Google Scholar 

  42. LaFave, L. M. et al. Reply to “Uveal melanoma cells are resistant to EZH2 inhibition regardless of BAP1 status”. Nat. Med. 22, 578–579 (2016).

    Article  PubMed  CAS  Google Scholar 

  43. LaFave, L. M. et al. Loss of BAP1 function leads to EZH2-dependent transformation. Nat. Med. 21, 1344–1349 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Schoumacher, M. et al. Uveal melanoma cells are resistant to EZH2 inhibition regardless of BAP1 status. Nat. Med. 22, 577–578 (2016).

    Article  PubMed  CAS  Google Scholar 

  45. Takahashi, Y. H. et al. Structural analysis of the core COMPASS family of histone H3K4 methylases from yeast to human. Proc. Natl Acad. Sci. USA 108, 20526–20531 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. McDonald, E. R. et al. Project DRIVE: a compendium of cancer dependencies and synthetic lethal relationships uncovered by large-scale, deep RNAi screening. Cell 170, 577–592 (2017).

    Article  PubMed  CAS  Google Scholar 

  47. Toska, E. et al. PI3K pathway regulates ER-dependent transcription in breast cancer through the epigenetic regulator KMT2D. Science 355, 1324–1330 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Lu, Z. H. et al. Mammalian target of rapamycin activator RHEB is frequently overexpressed in human carcinomas and is critical and sufficient for skin epithelial carcinogenesis. Cancer Res. 70, 3287–3298 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

  50. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Wang, L. et al. A cytoplasmic COMPASS is necessary for cell survival and triple-negative breast cancer pathogenesis by regulating metabolism. Genes Dev. 31, 2056–2066 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  PubMed  CAS  Google Scholar 

  53. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  PubMed  CAS  Google Scholar 

  54. Saldanha, A. J. Java Treeview—extensible visualization of microarray data. Bioinformatics 20, 3246–3248 (2004).

    Article  PubMed  CAS  Google Scholar 

  55. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. 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  PubMed  PubMed Central  CAS  Google Scholar 

  57. Shen, L., Shao, N., Liu, X. & Nestler, E. ngs.plot: quick mining and visualization of next-generation sequencing data by integrating genomic databases. BMC Genomics 15, 284 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR–Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank F. Zhang (MIT) for the kind gifts of the Px330 and lentiCRISPR v2 vectors. L.W. is supported by the Training Program in Signal Transduction and Cancer (T32 CA070085). Z.Z. is supported by the Robert H. Lurie Comprehensive Cancer Center—Translational Bridge Program Fellowship in Lymphoma Research. E.R.S. is supported by NCI grant R50CA211428. Studies in J.N.S.'s laboratory are supported by NIDCD grant DC013805, and studies related to COMPASS in A.S.'s laboratory are supported by NCI's Outstanding Investigator Award R35CA197569.

Author information

Authors and Affiliations

Authors

Contributions

L.W. and A.S. designed the study; L.W. and Z.Z. performed the majority of the experiments and part of the analyses, and wrote the first draft of the manuscript; R.H., N.L., X.H., L.W. and A.S. designed the in vivo studies; R.H., N.L. and X.H. performed and analyzed the in vivo experiments; S.A.M. and E.J.R. generated and sequenced the NGS libraries; K.A.C. and J.N.S. performed the mass spectrometry experiments and analyzed the results; C.K.C. performed the initial bioinformatics analyses on the studies related to the role of BAP1 and MLL3 at enhancers; P.A.O. performed all of the other bioinformatics analyses; J.J.M. provided clinical supervision in the interpretation of data; L.Z. and D.F. performed clinical data analysis; P.N. helped with the UTX ChIP-seq; Y.T. performed size-exclusion chromatography; L.W., Z.Z., M.A.M., E.R.S. and A.S. revised the manuscript.

Corresponding author

Correspondence to Ali Shilatifard.

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.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–20

Reporting Summary

Supplementary Table 1

Raw data for mass spectrometry of MLL3-NTD purification

Supplementary Table 2

Raw data for MLL3 mutations in human cancers

Supplementary Table 3

Genes that are significantly regulated by MLL3, BAP1 and UTX

Supplementary Table 4

Genes that are upregulated by MLL3 for more than twofold

Supplementary Table 5

Genes that are involved in epithelial cell differentiation pathway

Supplementary Table 6

GSEA pathway analysis for genes upregulated by MLL3

Supplementary Table 7

Genes that are upregulated by MLL3 and BAP1 and GSEA pathway analysis

Supplementary Table 8

Genes that are up-regulated by MLL3, BAP1 and UTX and GSEA pathway analysis

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, L., Zhao, Z., Ozark, P.A. et al. Resetting the epigenetic balance of Polycomb and COMPASS function at enhancers for cancer therapy. Nat Med 24, 758–769 (2018). https://doi.org/10.1038/s41591-018-0034-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0034-6

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