HOMEPRODUCTSCOMPANYCONTACTFAQResearchDictionaryPharmaSign Up FREE or Login

Loss of H3K27 Trimethylation Promotes Radiotherapy Resistance in Medulloblastoma and Induces an Actionable Vulnerability to BET Inhibition.

Abstract
Medulloblastoma has been categorized into four subgroups based on genetic, epigenetic, and transcriptional profiling. Radiation is used for treating medulloblastoma regardless of the subgroup. A better understanding of the molecular pathways determining radiotherapy response could help improve medulloblastoma treatment. Here, we investigated the role of the EZH2 (enhancer of zeste 2 polycomb repressive complex 2 subunit)-dependent histone H3K27 trimethylation in radiotherapy response in medulloblastoma. The tumors in 47.2% of patients with group 3 and 4 medulloblastoma displayed H3K27me3 deficiency. Loss of H3K27me3 was associated with a radioresistant phenotype, high relapse rates, and poor overall survival. In H3K27me3-deficient medulloblastoma cells, an epigenetic switch from H3K27me3 to H3K27ac occurred at specific genomic loci, altering the transcriptional profile. The resulting upregulation of EPHA2 stimulated excessive activation of the prosurvival AKT signaling pathway, leading to radiotherapy resistance. Bromodomain and extraterminal motif (BET) inhibition overcame radiation resistance in H3K27me3-deficient medulloblastoma cells by suppressing H3K27ac levels, blunting EPHA2 overexpression, and mitigating excessive AKT signaling. In addition, BET inhibition sensitized medulloblastoma cells to radiation by enhancing the apoptotic response through suppression of Bcl-xL and upregulation of Bim. This work demonstrates a novel mechanism of radiation resistance in medulloblastoma and identifies an epigenetic marker predictive of radiotherapy response. On the basis of these findings, we propose an epigenetically guided treatment approach targeting radiotherapy resistance in patients with medulloblastoma.
SIGNIFICANCE:
This study demonstrates a novel epigenetic mechanism of radiation resistance in medulloblastoma and identifies a therapeutic approach to improve outcomes in these patients.
AuthorsNishanth Gabriel, Kumaresh Balaji, Kay Jayachandran, Matthew Inkman, Jin Zhang, Sonika Dahiya, Michael Goldstein
JournalCancer research (Cancer Res) Vol. 82 Issue 10 Pg. 2019-2030 (05 16 2022) ISSN: 1538-7445 [Electronic] United States
PMID35315927 (Publication Type: Journal Article, Research Support, N.I.H., Extramural, Research Support, Non-U.S. Gov't)
Copyright©2022 American Association for Cancer Research.
Chemical References
  • DNER protein, human
  • Histones
  • Nerve Tissue Proteins
  • Receptors, Cell Surface
  • EZH2 protein, human
  • Enhancer of Zeste Homolog 2 Protein
  • Proto-Oncogene Proteins c-akt
Topics
  • Cerebellar Neoplasms (genetics, metabolism, pathology, radiotherapy)
  • Enhancer of Zeste Homolog 2 Protein (genetics, metabolism)
  • Histones (metabolism)
  • Humans
  • Medulloblastoma (genetics, metabolism, pathology, radiotherapy)
  • Neoplasm Recurrence, Local
  • Nerve Tissue Proteins (antagonists & inhibitors, genetics, metabolism)
  • Proto-Oncogene Proteins c-akt (metabolism)
  • Radiation Tolerance
  • Receptors, Cell Surface (antagonists & inhibitors, genetics, metabolism)

Join CureHunter, for free Research Interface BASIC access!

Take advantage of free CureHunter research engine access to explore the best drug and treatment options for any disease. Find out why thousands of doctors, pharma researchers and patient activists around the world use CureHunter every day.
Realize the full power of the drug-disease research graph!


Choose Username:
Email:
Password:
Verify Password:
Enter Code Shown: