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:

A chemical probe selectively inhibits G9a and GLP methyltransferase activity in cells

An Erratum to this article was published on 17 August 2011

Abstract

Protein lysine methyltransferases G9a and GLP modulate the transcriptional repression of a variety of genes via dimethylation of Lys9 on histone H3 (H3K9me2) as well as dimethylation of non-histone targets. Here we report the discovery of UNC0638, an inhibitor of G9a and GLP with excellent potency and selectivity over a wide range of epigenetic and non-epigenetic targets. UNC0638 treatment of a variety of cell lines resulted in lower global H3K9me2 levels, equivalent to levels observed for small hairpin RNA knockdown of G9a and GLP with the functional potency of UNC0638 being well separated from its toxicity. UNC0638 markedly reduced the clonogenicity of MCF7 cells, reduced the abundance of H3K9me2 marks at promoters of known G9a-regulated endogenous genes and disproportionately affected several genomic loci encoding microRNAs. In mouse embryonic stem cells, UNC0638 reactivated G9a-silenced genes and a retroviral reporter gene in a concentration-dependent manner without promoting differentiation.

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

Figure 1: UNC0638 competes with the peptide substrate but not with the cofactor SAM.
Figure 2: UNC0638 inhibits cellular H3K9 dimethylation and shows good separation of functional potency and toxicity in MDA-MB-231 cells.
Figure 3: Quantitative MS analysis of histone post-translational modifications in MDA-MB-231 cells.
Figure 4: Effects of UNC0638 on H3K9me2 and DNA methylation.
Figure 5: UNC0638 reactivates a silent EGFP retrovirus vector and G9a-regulated endogenous genes in mES cells.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

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

    Article  CAS  Google Scholar 

  2. Martin, C. & Zhang, Y. The diverse functions of histone lysine methylation. Nat. Rev. Mol. Cell Biol. 6, 838–849 (2005).

    Article  CAS  Google Scholar 

  3. Jenuwein, T. & Allis, C.D. Translating the histone code. Science 293, 1074–1080 (2001).

    Article  CAS  Google Scholar 

  4. Bernstein, B.E., Meissner, A. & Lander, E.S. The mammalian epigenome. Cell 128, 669–681 (2007).

    Article  CAS  Google Scholar 

  5. Gelato, K.A. & Fischle, W. Role of histone modifications in defining chromatin structure and function. Biol. Chem. 389, 353–363 (2008).

    Article  CAS  Google Scholar 

  6. Strahl, B.D. & Allis, C.D. The language of covalent histone modifications. Nature 403, 41–45 (2000).

    Article  CAS  Google Scholar 

  7. Huang, J. et al. Repression of p53 activity by Smyd2-mediated methylation. Nature 444, 629–632 (2006).

    Article  CAS  Google Scholar 

  8. Huang, J. et al. G9A and GLP methylate lysine 373 in the tumor suppressor p53. J. Biol. Chem. 285, 9636–9641 (2010).

    Article  CAS  Google Scholar 

  9. Rathert, P. et al. Protein lysine methyltransferase G9a acts on non-histone targets. Nat. Chem. Biol. 4, 344–346 (2008).

    Article  CAS  Google Scholar 

  10. Huang, J. et al. p53 is regulated by the lysine demethylase LSD1. Nature 449, 105–108 (2007).

    Article  CAS  Google Scholar 

  11. Copeland, R.A., Solomon, M.E. & Richon, V.M. Protein methyltransferases as a target class for drug discovery. Nat. Rev. Drug Discov. 8, 724–732 (2009).

    Article  CAS  Google Scholar 

  12. Rea, S. et al. Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature 406, 593–599 (2000).

    Article  CAS  Google Scholar 

  13. Cole, P.A. Chemical probes for histone-modifying enzymes. Nat. Chem. Biol. 4, 590–597 (2008).

    Article  CAS  Google Scholar 

  14. Esteller, M. Epigenetics in cancer. N. Engl. J. Med. 358, 1148–1159 (2008).

    Article  CAS  Google Scholar 

  15. Tachibana, M. et al. G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev. 16, 1779–1791 (2002).

    Article  CAS  Google Scholar 

  16. Kondo, Y. et al. Downregulation of histone H3 lysine 9 methyltransferase G9a induces centrosome disruption and chromosome instability in cancer cells. PLoS ONE 3, e2037 (2008).

    Article  Google Scholar 

  17. Kondo, Y. et al. Alterations of DNA methylation and histone modifications contribute to gene silencing in hepatocellular carcinomas. Hepatol. Res. 37, 974–983 (2007).

    Article  CAS  Google Scholar 

  18. Watanabe, H. et al. Deregulation of histone lysine methyltransferases contributes to oncogenic transformation of human bronchoepithelial cells. Cancer Cell Int. 8, 15 (2008).

    Article  Google Scholar 

  19. Goyama, S. et al. EVI-1 interacts with histone methyltransferases SUV39H1 and G9a for transcriptional repression and bone marrow immortalization. Leukemia 24, 81–88 (2010).

    Article  CAS  Google Scholar 

  20. Tachibana, M. et al. Histone methyltransferases G9a and GLP form heteromeric complexes and are both crucial for methylation of euchromatin at H3–K9. Genes Dev. 19, 815–826 (2005).

    Article  CAS  Google Scholar 

  21. Maze, I. et al. Essential role of the histone methyltransferase G9a in cocaine-induced plasticity. Science 327, 213–216 (2010).

    Article  CAS  Google Scholar 

  22. Schaefer, A. et al. Control of cognition and adaptive behavior by the GLP/G9a epigenetic suppressor complex. Neuron 64, 678–691 (2009).

    Article  CAS  Google Scholar 

  23. Imai, K., Togami, H. & Okamoto, T. Involvement of histone H3 Lysine 9 (H3K9) methyl transferase G9a in the maintenance of HIV-1 latency and its reactivation by BIX01294. J. Biol. Chem. 285, 16538–16545 (2010).

    Article  CAS  Google Scholar 

  24. Link, P.A. et al. Distinct roles for histone methyltransferases G9a and GLP in cancer germ-line antigen gene regulation in human cancer cells and murine embryonic stem cells. Mol. Cancer Res. 7, 851–862 (2009).

    Article  CAS  Google Scholar 

  25. Tachibana, M., Matsumura, Y., Fukuda, M., Kimura, H. & Shinkai, Y. G9a/GLP complexes independently mediate H3K9 and DNA methylation to silence transcription. EMBO J. 27, 2681–2690 (2008).

    Article  CAS  Google Scholar 

  26. Dong, K.B. et al. DNA methylation in ES cells requires the lysine methyltransferase G9a but not its catalytic activity. EMBO J. 27, 2691–2701 (2008).

    Article  CAS  Google Scholar 

  27. Shi, Y. et al. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell Stem Cell 3, 568–574 (2008).

    Article  CAS  Google Scholar 

  28. Shi, Y. et al. A combined chemical and genetic approach for the generation of induced pluripotent stem cells. Cell Stem Cell 2, 525–528 (2008).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. Frye, S.V. The art of the chemical probe. Nat. Chem. Biol. 6, 159–161 (2010).

    Article  CAS  Google Scholar 

  31. Liu, F. et al. Discovery of a 2,4-diamino-7-aminoalkoxyquinazoline as a potent and selective inhibitor of histone lysine methyltransferase G9a. J. Med. Chem. 52, 7950–7953 (2009).

    Article  CAS  Google Scholar 

  32. Liu, F. et al. Protein lysine methyltransferase G9a inhibitors: design, synthesis, and structure activity relationships of 2,4-diamino-7-aminoalkoxy-quinazolines. J. Med. Chem. 53, 5844–5857 (2010).

    Article  CAS  Google Scholar 

  33. Chang, Y. et al. Adding a lysine mimic in the design of potent inhibitors of histone lysine methyltransferases. J. Mol. Biol. 400, 1–7 (2010).

    Article  CAS  Google Scholar 

  34. Chang, Y. et al. Structural basis for G9a-like protein lysine methyltransferase inhibition by BIX-01294. Nat. Struct. Mol. Biol. 16, 312–317 (2009).

    Article  CAS  Google Scholar 

  35. Collazo, E., Couture, J.F., Bulfer, S. & Trievel, R.C. A coupled fluorescent assay for histone methyltransferases. Anal. Biochem. 342, 86–92 (2005).

    Article  CAS  Google Scholar 

  36. Wigle, T.J. et al. Accessing protein methyltransferase and demethylase enzymology using microfluidic capillary electrophoresis. Chem. Biol. 17, 695–704 (2010).

    Article  CAS  Google Scholar 

  37. Morrison, J.F. Kinetics of the reversible inhibition of enzyme-catalysed reactions by tight-binding inhibitors. Biochim. Biophys. Acta 185, 269–286 (1969).

    Article  CAS  Google Scholar 

  38. Zee, B.M. et al. In vivo residue-specific histone methylation dynamics. J. Biol. Chem. 285, 3341–3350 (2010).

    Article  CAS  Google Scholar 

  39. Plazas-Mayorca, M.D. et al. Quantitative proteomics reveals direct and indirect alterations in the histone code following methyltransferase knockdown. Mol. Biosyst. 6, 1719–1729 (2010).

    Article  CAS  Google Scholar 

  40. Barski, A. et al. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837 (2007).

    Article  CAS  Google Scholar 

  41. Rice, J.C. et al. Histone methyltransferases direct different degrees of methylation to define distinct chromatin domains. Mol. Cell 12, 1591–1598 (2003).

    Article  CAS  Google Scholar 

  42. Vakoc, C.R., Mandat, S.A., Olenchock, B.A. & Blobel, G.A. Histone H3 lysine 9 methylation and HP1gamma are associated with transcription elongation through mammalian chromatin. Mol. Cell 19, 381–391 (2005).

    Article  CAS  Google Scholar 

  43. Wolf, D. & Goff, S.P. TRIM28 mediates primer binding site-targeted silencing of murine leukemia virus in embryonic cells. Cell 131, 46–57 (2007).

    Article  CAS  Google Scholar 

  44. Yokochi, T. et al. G9a selectively represses a class of late-replicating genes at the nuclear periphery. Proc. Natl. Acad. Sci. USA 106, 19363–19368 (2009).

    Article  CAS  Google Scholar 

  45. Hosey, A.M., Chaturvedi, C.P. & Brand, M. Crosstalk between histone modifications maintains the developmental pattern of gene expression on a tissue-specific locus. Epigenetics 5, 273–281 (2010).

    Article  CAS  Google Scholar 

  46. Epsztejn-Litman, S. et al. De novo DNA methylation promoted by G9a prevents reprogramming of embryonically silenced genes. Nat. Struct. Mol. Biol. 15, 1176–1183 (2008).

    Article  CAS  Google Scholar 

  47. Estève, P.O. et al. Direct interaction between DNMT1 and G9a coordinates DNA and histone methylation during replication. Genes Dev. 20, 3089–3103 (2006).

    Article  Google Scholar 

  48. Chen, L. et al. MDM2 recruitment of lysine methyltransferases regulates p53 transcriptional output. EMBO J. 29, 2538–2552 (2010).

    Article  CAS  Google Scholar 

  49. Fritsch, L. et al. A subset of the histone H3 lysine 9 methyltransferases Suv39h1, G9a, GLP, and SETDB1 participate in a multimeric complex. Mol. Cell 37, 46–56 (2010).

    Article  CAS  Google Scholar 

  50. Stadtfeld, M., Maherali, N., Breault, D.T. & Hochedlinger, K. Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse. Cell Stem Cell 2, 230–240 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Tumber for JMJD2E assay support; J. Moffat (University of Toronto) for the gift of shRNAs; R. Bristow (University Health Network) for RV221 and PC3 cells; T. Hajian and F. Syeda for protein purification; G. Senisterra for contributing to DSF and DSLS data analysis; M. Herold for graphical design and illustration; I. Korboukh, M. Herold and J. Yost for critical reading of the manuscript; and R. Trump and C. Yates for helpful discussion. The research described here was supported by the National Institute of General Medical Sciences, US National Institutes of Health (NIH; grant RC1GM090732), the Carolina Partnership and University Cancer Research Fund from the University of North Carolina at Chapel Hill, the US National Science Foundation (NSF), the Ontario Research Fund, the Ontario Ministry of Health and Long-term Care and the Structural Genomics Consortium. The Structural Genomics Consortium is a registered charity (number 1097737) that receives funds from the Canadian Institutes for Health Research (CIHR), the Canada Foundation for Innovation, Genome Canada through the Ontario Genomics Institute, GlaxoSmithKline, Karolinska Institutet, the Knut and Alice Wallenberg Foundation, the Ontario Innovation Trust, the Ontario Ministry for Research and Innovation, Merck & Co. Inc., the Novartis Research Foundation, the Swedish Agency for Innovation Systems, the Swedish Foundation for Strategic Research and the Wellcome Trust. C.H.A. holds a Canada Research Chair in Structural Genomics. V.L. is supported by a CIHR fellowship. A.P. is supported by grants from the CIHR (199170 and 186007) and from the NIH (MH074127, MH088413, DP3DK085698 and HG004535). A.P. is Tapscott Chair in Schizophrenia Studies and a Senior Fellow of the Ontario Mental Health Foundation. J.E. is supported by CIHR grant IG1-102956. B.A.G. is supported by grants from the NSF (Early Faculty CAREER award and CBET-0941143), the American Society for Mass Spectrometry and the NIH Office of the Director (DP2OD007447). P.A.D. is supported by NIH postdoctoral fellowship F32 NRSA.

Author information

Authors and Affiliations

Authors

Contributions

M.V., A.A.-H., A.S. and I.C. performed SAHH-coupled, fluorescence-polarization, DSF, DSLS and DNMT1 assays; D.B.-L. developed and performed in-cell western, MTT, ChIP, gene expression, clonogenicity, western blotting and immunofluorescence studies; F.L. developed the synthetic route to UNC0638 and UNC0737 and synthesized the compounds; S.R.-G. and J.E. developed and performed mES cell studies; V.L., S.-C.W. and A.P. performed H3K9me2 genomic localization and DNA methylation analysis; T.J.W. and W.P.J. performed mechanism-of-action studies; P.A.D. and B.A.G. performed MS-based proteomics studies; M.V., G.A.W., A.D., W.T., D.B.K. and C.H.A. solved and analyzed the X-ray crystal structure of the G9a-UNC0638-SAH complex; T.J.W. and A.T. performed SPR studies; X.C. and S.G.P. performed UNC0638 stability studies; S.G.P. performed RT-qPCR studies; T.J.M., X.-p.H. and B.L.R. performed GPCR selectivity studies; C.D.S. and W.P.J. performed kinase selectivity studies; J.L.N. purified proteins; J.J. designed UNC0638 and UNC0737; C.H.A., J.J., S.V.F., M.V., D.B.-L., P.J.B., J.E., S.R.-G., A.P., V.L., B.A.G., T.J.W. and A.E. designed studies and discussed results; J.J., C.H.A., D.B.-L., S.R.-G., M.V., V.L., S.V.F., J.E., A.P., B.A.G., P.J.B. and T.J.W. wrote the paper.

Corresponding authors

Correspondence to Cheryl H Arrowsmith or Jian Jin.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Methods and Supplementary Results (PDF 2603 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Vedadi, M., Barsyte-Lovejoy, D., Liu, F. et al. A chemical probe selectively inhibits G9a and GLP methyltransferase activity in cells. Nat Chem Biol 7, 566–574 (2011). https://doi.org/10.1038/nchembio.599

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.599

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