Reduced H3K27me3 and DNA hypomethylation are major drivers of gene expression in K27M mutant pediatric high-grade gliomas

Research output: Contribution to journalJournal articleResearchpeer-review

  • Sebastian Bender
  • Yujie Tang
  • Anders M Lindroth
  • Volker Hovestadt
  • David T W Jones
  • Marcel Kool
  • Marc Zapatka
  • Paul A Northcott
  • Dominik Sturm
  • Wei Guo Wang
  • Bernhard Radlwimmer
  • Jonas W Højfeldt
  • Nathalène Truffaux
  • David Castel
  • Simone Schubert
  • Marina Ryzhova
  • Huriye Seker-Cin
  • Jan Gronych
  • Pascal David Johann
  • Sebastian Stark
  • And 21 others
  • Jochen Meyer
  • Till Milde
  • Martin Schuhmann
  • Martin Ebinger
  • Camelia-Maria Monoranu
  • Anitha Ponnuswami
  • Spenser Chen
  • Chris Jones
  • Olaf Witt
  • V Peter Collins
  • Andreas von Deimling
  • Nada Jabado
  • Stephanie Puget
  • Jacques Grill
  • Kristian Helin
  • Andrey Korshunov
  • Peter Lichter
  • Michelle Monje
  • Christoph Plass
  • Yoon-Jae Cho
  • Stefan M Pfister
Two recurrent mutations, K27M and G34R/V, within histone variant H3.3 were recently identified in ∼50% of pHGGs. Both mutations define clinically and biologically distinct subgroups of pHGGs. Here, we provide further insight about the dominant-negative effect of K27M mutant H3.3, leading to a global reduction of the repressive histone mark H3K27me3. We demonstrate that this is caused by aberrant recruitment of the PRC2 complex to K27M mutant H3.3 and enzymatic inhibition of the H3K27me3-establishing methyltransferase EZH2. By performing chromatin immunoprecipitation followed by next-generation sequencing and whole-genome bisulfite sequencing in primary pHGGs, we show that reduced H3K27me3 levels and DNA hypomethylation act in concert to activate gene expression in K27M mutant pHGGs.
Original languageEnglish
JournalCancer Cell
Volume24
Issue number5
Pages (from-to)660-72
Number of pages13
ISSN1535-6108
DOIs
Publication statusPublished - 11 Nov 2013

ID: 94394591