Your browser doesn't support javascript.
loading
EZH2 mediated metabolic rewiring promotes tumor growth independently of histone methyltransferase activity in ovarian cancer.
Chen, Jianfeng; Hong, Jing Han; Huang, Yulin; Liu, Shini; Yin, Jiaxin; Deng, Peng; Sun, Yichen; Yu, Zhaoliang; Zeng, Xian; Xiao, Rong; Chan, Jason Yongsheng; Guan, Peiyong; Wang, Yali; Wang, Peili; Liu, Lizhen; Wen, Shijun; Yu, Qiang; Ong, Choon Kiat; Teh, Bin-Tean; Xiong, Ying; Tan, Jing.
Affiliation
  • Chen J; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China. chenjf@sysucc.org.cn.
  • Hong JH; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou, Guangdong, 510060, P. R. China. chenjf@sysucc.org.cn.
  • Huang Y; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore.
  • Liu S; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
  • Yin J; Department of Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510060, P. R. China.
  • Deng P; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
  • Sun Y; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
  • Yu Z; Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangdong, 510060, P. R. China.
  • Zeng X; Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, P. R. China.
  • Xiao R; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
  • Chan JY; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
  • Guan P; Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore.
  • Wang Y; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore.
  • Wang P; Genome Institute of Singapore, A*STAR, Singapore, Singapore.
  • Liu L; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
  • Wen S; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
  • Yu Q; Center of Medical Research, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510060, P. R. China.
  • Ong CK; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
  • Teh BT; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore.
  • Xiong Y; Genome Institute of Singapore, A*STAR, Singapore, Singapore.
  • Tan J; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore.
Mol Cancer ; 22(1): 85, 2023 05 20.
Article in En | MEDLINE | ID: mdl-37210576
ABSTRACT

BACKGROUND:

Enhancer of zeste homolog 2 (EZH2), the key catalytic subunit of polycomb repressive complex 2 (PRC2), is overexpressed and plays an oncogenic role in various cancers through catalysis-dependent or catalysis-independent pathways. However, the related mechanisms contributing to ovarian cancer (OC) are not well understood.

METHODS:

The levels of EZH2 and H3K27me3 were evaluated in 105 OC patients by immunohistochemistry (IHC) staining, and these patients were stratified based on these levels. Canonical and noncanonical binding sites of EZH2 were defined by chromatin immunoprecipitation sequencing (ChIP-Seq). The EZH2 solo targets were obtained by integrative analysis of ChIP-Seq and RNA sequencing data. In vitro and in vivo experiments were performed to determine the role of EZH2 in OC growth.

RESULTS:

We showed that a subgroup of OC patients with high EZH2 expression but low H3K27me3 exhibited the worst prognosis, with limited therapeutic options. We demonstrated that induction of EZH2 degradation but not catalytic inhibition profoundly blocked OC cell proliferation and tumorigenicity in vitro and in vivo. Integrative analysis of genome-wide chromatin and transcriptome profiles revealed extensive EZH2 occupancy not only at genomic loci marked by H3K27me3 but also at promoters independent of PRC2, indicating a noncanonical role of EZH2 in OC. Mechanistically, EZH2 transcriptionally upregulated IDH2 to potentiate metabolic rewiring by enhancing tricarboxylic acid cycle (TCA cycle) activity, which contributed to the growth of OC.

CONCLUSIONS:

These data reveal a novel oncogenic role of EZH2 in OC and identify potential therapeutic strategies for OC by targeting the noncatalytic activity of EZH2.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Ovarian Neoplasms / Enhancer of Zeste Homolog 2 Protein Limits: Female / Humans Language: En Journal: Mol Cancer Journal subject: NEOPLASIAS Year: 2023 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Ovarian Neoplasms / Enhancer of Zeste Homolog 2 Protein Limits: Female / Humans Language: En Journal: Mol Cancer Journal subject: NEOPLASIAS Year: 2023 Document type: Article