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.

  • Letter
  • Published:

Therapeutic targeting of polycomb and BET bromodomain proteins in diffuse intrinsic pontine gliomas

Abstract

Diffuse intrinsic pontine glioma (DIPG) is a highly aggressive pediatric brainstem tumor characterized by rapid and uniform patient demise1. A heterozygous point mutation of histone H3 occurs in more than 80% of these tumors and results in a lysine-to-methionine substitution (H3K27M)2,3. Expression of this histone mutant is accompanied by a reduction in the levels of polycomb repressive complex 2 (PRC2)-mediated H3K27 trimethylation (H3K27me3), and this is hypothesized to be a driving event of DIPG oncogenesis4,5. Despite a major loss of H3K27me3, PRC2 activity is still detected in DIPG cells positive for H3K27M6,7. To investigate the functional roles of H3K27M and PRC2 in DIPG pathogenesis, we profiled the epigenome of H3K27M-mutant DIPG cells and found that H3K27M associates with increased H3K27 acetylation (H3K27ac). In accordance with previous biochemical data5, the majority of the heterotypic H3K27M-K27ac nucleosomes colocalize with bromodomain proteins at the loci of actively transcribed genes, whereas PRC2 is excluded from these regions; this suggests that H3K27M does not sequester PRC2 on chromatin. Residual PRC2 activity is required to maintain DIPG proliferative potential, by repressing neuronal differentiation and function. Finally, to examine the therapeutic potential of blocking the recruitment of bromodomain proteins by heterotypic H3K27M-K27ac nucleosomes in DIPG cells, we performed treatments in vivo with BET bromodomain inhibitors and demonstrate that they efficiently inhibit tumor progression, thus identifying this class of compounds as potential therapeutics in DIPG.

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

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

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

Figure 1: H3K27M correlates with H3K27ac and is excluded from PRC2 targets.
Figure 2: PRC2 is required for the oncogenic potential of H3K27M DIPG cells.
Figure 3: Bromodomain-protein inhibition impairs proliferation and triggers differentiation of H3K27M-positive DIPG cells.
Figure 4: Bromodomain-protein inhibition significantly extends survival of DIPG xenograft model.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Schroeder, K.M., Hoeman, C.M. & Becher, O.J. Children are not just little adults: recent advances in understanding of diffuse intrinsic pontine glioma biology. Pediatr. Res. 75, 205–209 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Schwartzentruber, J. et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482, 226–231 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Wu, G. et al. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat. Genet. 44, 251–253 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lewis, P.W. et al. Inhibition of PRC2 activity by a gain-of-function H3 mutation found in pediatric glioblastoma. Science 340, 857–861 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bender, S. et al. Reduced H3K27me3 and DNA hypomethylation are major drivers of gene expression in K27M mutant pediatric high-grade gliomas. Cancer Cell 24, 660–672 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Chan, K.M. et al. The histone H3.3K27M mutation in pediatric glioma reprograms H3K27 methylation and gene expression. Genes Dev. 27, 985–990 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hashizume, R. et al. Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma. Nat. Med. 20, 1394–1396 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Monje, M. et al. Hedgehog-responsive candidate cell of origin for diffuse intrinsic pontine glioma. Proc. Natl. Acad. Sci. USA 108, 4453–4458 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Justin, N. et al. Structural basis of oncogenic histone H3K27M inhibition of human polycomb repressive complex 2. Nat. Commun. 7, 11316 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  12. Bracken, A.P. et al. The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells. Genes Dev. 21, 525–530 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Piunti, A. et al. Polycomb proteins control proliferation and transformation independently of cell cycle checkpoints by regulating DNA replication. Nat. Commun. 5, 3649 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Qi, J. Bromodomain and extraterminal domain inhibitors (BETi) for cancer therapy: chemical modulation of chromatin structure. Cold Spring Harb. Perspect. Biol. 6, a018663 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Jonkers, I. & Lis, J.T. Getting up to speed with transcription elongation by RNA polymerase II. Nat. Rev. Mol. Cell Biol. 16, 167–177 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Galderisi, U., Jori, F.P. & Giordano, A. Cell cycle regulation and neural differentiation. Oncogene 22, 5208–5219 (2003).

    Article  CAS  PubMed  Google Scholar 

  18. Gordon, J., Amini, S. & White, M.K. General overview of neuronal cell culture. Methods Mol. Biol. 1078, 1–8 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. An, W., Palhan, V.B., Karymov, M.A., Leuba, S.H. & Roeder, R.G. Selective requirements for histone H3 and H4 N termini in p300-dependent transcriptional activation from chromatin. Mol. Cell 9, 811–821 (2002).

    Article  CAS  PubMed  Google Scholar 

  20. Filippakopoulos, P. & Knapp, S. Targeting bromodomains: epigenetic readers of lysine acetylation. Nat. Rev. Drug Discov. 13, 337–356 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Gupta, S., Takebe, N. & Lorusso, P. Targeting the Hedgehog pathway in cancer. Ther. Adv. Med. Oncol. 2, 237–250 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Whitfield, M.L., George, L.K., Grant, G.D. & Perou, C.M. Common markers of proliferation. Nat. Rev. Cancer 6, 99–106 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Weiner, A. et al. Co-ChIP enables genome-wide mapping of histone mark co-occurrence at single-molecule resolution. Nat. Biotechnol. 34, 953–961 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Pasini, D. et al. Characterization of an antagonistic switch between histone H3 lysine 27 methylation and acetylation in the transcriptional regulation of Polycomb group target genes. Nucleic Acids Res. 38, 4958–4969 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ferrari, K.J. et al. Polycomb-dependent H3K27me1 and H3K27me2 regulate active transcription and enhancer fidelity. Mol. Cell 53, 49–62 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Mohn, F. et al. Lineage-specific polycomb targets and de novo DNA methylation define restriction and potential of neuronal progenitors. Mol. Cell 30, 755–766 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Hanahan, D. & Weinberg, R.A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Jones, C. & Baker, S.J. Unique genetic and epigenetic mechanisms driving paediatric diffuse high-grade glioma. Nat. Rev. Cancer 14, 651–661 (2014).

    Article  CAS  Google Scholar 

  29. Grasso, C.S. et al. Functionally defined therapeutic targets in diffuse intrinsic pontine glioma. Nat. Med. 21, 827 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Mazur, P.K. et al. Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinoma. Nat. Med. 21, 1163–1171 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Bhadury, J. et al. BET and HDAC inhibitors induce similar genes and biological effects and synergize to kill in Myc-induced murine lymphoma. Proc. Natl. Acad. Sci. USA 111, E2721–E2730 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hashizume, R. et al. Characterization of a diffuse intrinsic pontine glioma cell line: implications for future investigations and treatment. J. Neurooncol. 110, 305–313 (2012).

    Article  PubMed  Google Scholar 

  33. Mueller, S. et al. Targeting Wee1 for the treatment of pediatric high-grade gliomas. Neuro-oncol. 16, 352–360 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Chen, F.X. et al. PAF1, a molecular regulator of promoter-proximal pausing by RNA polymerase II. Cell 162, 1003–1015 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chen, F., Gao, X. & Shilatifard, A. Stably paused genes revealed through inhibition of transcription initiation by the TFIIH inhibitor triptolide. Genes Dev. 29, 39–47 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Lee, T.I., Johnstone, S.E. & Young, R.A. Chromatin immunoprecipitation and microarray-based analysis of protein location. Nat. Protoc. 1, 729–748 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bolger, A.M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Zang, C. et al. A clustering approach for identification of enriched domains from histone modification ChIP-Seq data. Bioinformatics 25, 1952–1958 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  41. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  43. 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  CAS  PubMed  Google Scholar 

  44. Meerbrey, K.L. et al. The pINDUCER lentiviral toolkit for inducible RNA interference in vitro and in vivo. Proc. Natl. Acad. Sci. USA 108, 3665–3670 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all the members of the Shilatifard lab for their useful comments and suggestions. We thank M. Monje (Stanford University) for use of the SU-DIPG-IV cell line. We thank D. Pasini (European Institute of Oncology, IEO) for providing the SUZ12 and control vectors, and C. Rivetta for technical support. pInducer20 was a gift from S. Elledge (deposited in Addgene, plasmid #44012). A.P. is supported by a long-term EMBO fellowship (ALTF 372-2015), and his work in the Shilatifard lab is supported by AIRC and Marie Curie Actions—People—COFUND. R.H. is supported by US National Institutes of Health grant RO1NS093079, the Matthew Larson Foundation and the Bear Necessities Pediatric Cancer Foundation and Rally Foundation. Proteomics services were performed by the Northwestern Proteomics Core Facility, generously supported by NCI CCSG P30CA060553 awarded to the Robert H. Lurie Comprehensive Cancer Center, and the National Resource for Translational and Developmental Proteomics supported by P41GM108569. Studies in regards to the development of targeted therapeutics for DIPG within our groups are partially supported by the generous support of J. McNicholas Pediatric Brain Tumor Foundation. Studies in the Shilatifard laboratory are supported by NCI grant R35CA197569.

Author information

Authors and Affiliations

Authors

Contributions

A.P. and A.S. designed the study. A.P. performed the majority of the experiments, part of the analyses and wrote the manuscript. R.H., C.D.J., A.P. and A.S. designed the in vivo studies. R.H. and Q.M. performed and analyzed the in vivo experiments. M.A.M. performed the MNase-IP experiment. A.R.W. performed the initial bioinformatics analyses on the studies related to the role of PRC2 in DIPG. E.T.B. performed all other bioinformatics analyses. C.M.H. performed and analyzed the immunohistochemistry studies. S.A.M. and E.J.R. generated and sequenced the next-generation sequencing (NGS) libraries. Y.-h.T. provided technical help. A.V.M.performed the FACS studies. N.A.A. and N.L.K. designed the mass spectrometry studies. N.A.A. performed the mass spectrometry experiments. R.R.L. and A.M.S. provided clinical supervision in the interpretation of data. A.P., M.A.M., C.D.J. and A.S. revised the manuscript. All authors commented on the manuscript and approved data included in it.

Corresponding authors

Correspondence to C David James or Ali Shilatifard.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text & Figures

Supplementary Methods & Supplementary Figures 1–18 (PDF 6282 kb)

Supplementary Table 1

Mass spectrometry data (XLSX 41 kb)

Supplementary Table 2

Antibody's list (XLSX 10 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Piunti, A., Hashizume, R., Morgan, M. et al. Therapeutic targeting of polycomb and BET bromodomain proteins in diffuse intrinsic pontine gliomas. Nat Med 23, 493–500 (2017). https://doi.org/10.1038/nm.4296

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4296

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