Cited time in webofscience Cited time in scopus

Epigenetically defined therapeutic targeting in H3.3G34R/V high-grade gliomas

Title
Epigenetically defined therapeutic targeting in H3.3G34R/V high-grade gliomas
Author(s)
Sweha, Stefan R.Chung, ChanNatarajan, Siva KumarPanwalkar, PoojaPun, MatthewGhali, AmerBayliss, JillPratt, DrewShankar, AnandRavikumar, VisweswaranRao, ArvindCieslik, MarcinWilder-Romans, KariScott, Andrew J.Wahl, Daniel R.Jessa, SelinKleinman, Claudia L.Jabado, NadaMackay, AlanJones, ChrisMartinez, DanielSanti, MariaritaJudkins, Alexander R.Yadav, Viveka NandQin, TingtingPhoenix, Timothy N.Koschmann, Carl J.Baker, Suzanne J.Chinnaiyan, Arul M.Venneti, Sriram
DGIST Authors
Sweha, Stefan R.Chung, ChanNatarajan, Siva KumarPanwalkar, PoojaPun, MatthewGhali, AmerBayliss, JillPratt, DrewShankar, AnandRavikumar, VisweswaranRao, ArvindCieslik, MarcinWilder-Romans, KariScott, Andrew J.Wahl, Daniel R.Jessa, SelinKleinman, Claudia L.Jabado, NadaMackay, AlanJones, ChrisMartinez, DanielSanti, MariaritaJudkins, Alexander R.Yadav, Viveka NandQin, TingtingPhoenix, Timothy N.Koschmann, Carl J.Baker, Suzanne J.Chinnaiyan, Arul M.Venneti, Sriram
Issued Date
2021-10
Type
Article
Keywords
CHROMATIN-STATE DISCOVERYCENTRAL-NERVOUS-SYSTEMPEDIATRIC HIGH-GRADESIGNAL TRANSDUCERSPRC2 ACTIVITYMUTATIONSINHIBITORMETHYLATIONPHOSPHORYLATIONPROGENITORS
ISSN
1946-6234
Abstract
High-grade gliomas with arginine or valine substitutions of the histone H3.3 glycine-34 residue (H3.3G34R/V) carry a dismal prognosis, and current treatments, including radiotherapy and chemotherapy, are not curative. Because H3.3G34R/V mutations reprogram epigenetic modifications, we undertook a comprehensive epigenetic approach using ChIP sequencing and ChromHMM computational analysis to define therapeutic dependencies in H3.3G34R/V gliomas. Our analyses revealed a convergence of epigenetic alterations, including (i) activating epigenetic modifications on histone H3 lysine (K) residues such as H3K36 trimethylation (H3K36me3), H3K27 acetylation (H3K27ac), and H3K4 trimethylation (H3K4me3); (ii) DNA promoter hypomethylation; and (iii) redistribution of repressive histone H3K27 trimethylation (H3K27me3) to intergenic regions at the leukemia inhibitory factor (LIF) locus to drive increased LIF abundance and secretion by H3.3G34R/V cells. LIF activated signal transducer and activator of transcription 3 (STAT3) signaling in an autocrine/paracrine manner to promote survival of H3.3G34R/V glioma cells. Moreover, immunohistochemistry and single-cell RNA sequencing from H3.3G34R/V patient tumors revealed high STAT3 protein and RNA expression, respectively, in tumor cells with both inter- and intratumor heterogeneity. We targeted STAT3 using a blood-brain barrier-penetrable small-molecule inhibitor, WP1066, currently in clinical trials for adult gliomas. WP1066 treatment resulted in H3.3G34R/V tumor cell toxicity in vitro and tumor suppression in preclinical mouse models established with KNS42 cells, SJ-HGGx42-c cells, or in utero electroporation techniques. Our studies identify the LIF/STAT3 pathway as a key epigenetically driven and druggable vulnerability in H3.3G34R/V gliomas. This finding could inform development of targeted, combination therapies for these lethal brain tumors.
URI
http://hdl.handle.net/20.500.11750/15756
DOI
10.1126/scitranslmed.abf7860
Publisher
American Association for the Advancement of Science
Related Researcher
  • 정찬 Chung, Chan
  • Research Interests Cancer Epigenetics; Lysosome; Neurodegeneration; Central Nervous System; Hepatitis; Lysosomal Storage Diseases; Endosomes; Cell Culture; Immunohistochemistry; Neurodegenerative Diseases; Brain Diseases
Files in This Item:

There are no files associated with this item.

Appears in Collections:
Department of New Biology Cancer Epigenetics Lab 1. Journal Articles

qrcode

  • twitter
  • facebook
  • mendeley

Items in Repository are protected by copyright, with all rights reserved, unless otherwise indicated.

BROWSE