Your browser doesn't support javascript.
loading
Histone deposition pathways determine the chromatin landscapes of H3.1 and H3.3 K27M oncohistones.
Sarthy, Jay F; Meers, Michael P; Janssens, Derek H; Henikoff, Jorja G; Feldman, Heather; Paddison, Patrick J; Lockwood, Christina M; Vitanza, Nicholas A; Olson, James M; Ahmad, Kami; Henikoff, Steven.
Affiliation
  • Sarthy JF; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, United States.
  • Meers MP; Cancer and Blood Disorders, Seattle, United States.
  • Janssens DH; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, United States.
  • Henikoff JG; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, United States.
  • Feldman H; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, United States.
  • Paddison PJ; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, United States.
  • Lockwood CM; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, United States.
  • Vitanza NA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, United States.
  • Olson JM; Cancer and Blood Disorders, Seattle, United States.
  • Ahmad K; Clinical Research Division Fred Hutchinson Cancer Research Center, Seattle, United States.
  • Henikoff S; Cancer and Blood Disorders, Seattle, United States.
Elife ; 92020 09 09.
Article in En | MEDLINE | ID: mdl-32902381
ABSTRACT
Lysine 27-to-methionine (K27M) mutations in the H3.1 or H3.3 histone genes are characteristic of pediatric diffuse midline gliomas (DMGs). These oncohistone mutations dominantly inhibit histone H3K27 trimethylation and silencing, but it is unknown how oncohistone type affects gliomagenesis. We show that the genomic distributions of H3.1 and H3.3 oncohistones in human patient-derived DMG cells are consistent with the DNAreplication-coupled deposition of histone H3.1 and the predominant replication-independent deposition of histone H3.3. Although H3K27 trimethylation is reduced for both oncohistone types, H3.3K27M-bearing cells retain some domains, and only H3.1K27M-bearing cells lack H3K27 trimethylation. Neither oncohistone interferes with PRC2 binding. Using Drosophila as a model, we demonstrate that inhibition of H3K27 trimethylation occurs only when H3K27M oncohistones are deposited into chromatin and only when expressed in cycling cells. We propose that oncohistones inhibit the H3K27 methyltransferase as chromatin patterns are being duplicated in proliferating cells, predisposing them to tumorigenesis.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Chromatin / Histones / Gene Expression Regulation, Neoplastic / Mutation Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Elife Year: 2020 Document type: Article Affiliation country: United States Publication country: ENGLAND / ESCOCIA / GB / GREAT BRITAIN / INGLATERRA / REINO UNIDO / SCOTLAND / UK / UNITED KINGDOM

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Chromatin / Histones / Gene Expression Regulation, Neoplastic / Mutation Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Elife Year: 2020 Document type: Article Affiliation country: United States Publication country: ENGLAND / ESCOCIA / GB / GREAT BRITAIN / INGLATERRA / REINO UNIDO / SCOTLAND / UK / UNITED KINGDOM