Your browser doesn't support javascript.
loading
Modeling epigenetic lesions that cause gliomas.
Rahme, Gilbert J; Javed, Nauman M; Puorro, Kaitlyn L; Xin, Shouhui; Hovestadt, Volker; Johnstone, Sarah E; Bernstein, Bradley E.
Affiliation
  • Rahme GJ; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Departments of Cell Biology and Pathology, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
  • Javed NM; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Departments of Cell Biology and Pathology, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
  • Puorro KL; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
  • Xin S; Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA.
  • Hovestadt V; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
  • Johnstone SE; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
  • Bernstein BE; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Departments of Cell Biology and Pathology, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA. Electronic address: bradley_bernstein@dfci.harvard.edu.
Cell ; 186(17): 3674-3685.e14, 2023 08 17.
Article in En | MEDLINE | ID: mdl-37494934
ABSTRACT
Epigenetic lesions that disrupt regulatory elements represent potential cancer drivers. However, we lack experimental models for validating their tumorigenic impact. Here, we model aberrations arising in isocitrate dehydrogenase-mutant gliomas, which exhibit DNA hypermethylation. We focus on a CTCF insulator near the PDGFRA oncogene that is recurrently disrupted by methylation in these tumors. We demonstrate that disruption of the syntenic insulator in mouse oligodendrocyte progenitor cells (OPCs) allows an OPC-specific enhancer to contact and induce Pdgfra, thereby increasing proliferation. We show that a second lesion, methylation-dependent silencing of the Cdkn2a tumor suppressor, cooperates with insulator loss in OPCs. Coordinate inactivation of the Pdgfra insulator and Cdkn2a drives gliomagenesis in vivo. Despite locus synteny, the insulator is CpG-rich only in humans, a feature that may confer human glioma risk but complicates mouse modeling. Our study demonstrates the capacity of recurrent epigenetic lesions to drive OPC proliferation in vitro and gliomagenesis in vivo.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Brain Neoplasms / Epigenesis, Genetic / Glioma Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Cell Year: 2023 Document type: Article Affiliation country:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Brain Neoplasms / Epigenesis, Genetic / Glioma Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Cell Year: 2023 Document type: Article Affiliation country:
...