Your browser doesn't support javascript.
loading
Histone demethylase KDM4C controls tumorigenesis of glioblastoma by epigenetically regulating p53 and c-Myc.
Lee, Dong Hoon; Kim, Go Woon; Yoo, Jung; Lee, Sang Wu; Jeon, Yu Hyun; Kim, So Yeon; Kang, Hyeok Gu; Kim, Da-Hyun; Chun, Kyung-Hee; Choi, Junjeong; Kwon, So Hee.
Affiliation
  • Lee DH; College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea.
  • Kim GW; College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea.
  • Yoo J; College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea.
  • Lee SW; College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea.
  • Jeon YH; College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea.
  • Kim SY; College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea.
  • Kang HG; Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Kim DH; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Chun KH; Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Choi J; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Kwon SH; Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
Cell Death Dis ; 12(1): 89, 2021 01 18.
Article in En | MEDLINE | ID: mdl-33462212
ABSTRACT
Glioblastoma is the most lethal brain tumor and its pathogenesis remains incompletely understood. KDM4C is a histone H3K9 demethylase that contributes to epigenetic regulation of both oncogene and tumor suppressor genes and is often overexpressed in human tumors, including glioblastoma. However, KDM4C's roles in glioblastoma and the underlying molecular mechanisms remain unclear. Here, we show that KDM4C knockdown significantly represses proliferation and tumorigenesis of glioblastoma cells in vitro and in vivo that are rescued by overexpressing wild-type KDM4C but not a catalytic dead mutant. KDM4C protein expression is upregulated in glioblastoma, and its expression correlates with c-Myc expression. KDM4C also binds to the c-Myc promoter and induces c-Myc expression. Importantly, KDM4C suppresses the pro-apoptotic functions of p53 by demethylating p53K372me1, which is pivotal for the stability of chromatin-bound p53. Conversely, depletion or inhibition of KDM4C promotes p53 target gene expression and induces apoptosis in glioblastoma. KDM4C may serve as an oncogene through the dual functions of inactivation of p53 and activation of c-Myc in glioblastoma. Our study demonstrates KDM4C inhibition as a promising therapeutic strategy for targeting glioblastoma.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Brain Neoplasms / Tumor Suppressor Protein p53 / Proto-Oncogene Proteins c-myc / Glioblastoma / Jumonji Domain-Containing Histone Demethylases Type of study: Clinical_trials Limits: Animals / Humans Language: En Journal: Cell Death Dis Year: 2021 Type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Brain Neoplasms / Tumor Suppressor Protein p53 / Proto-Oncogene Proteins c-myc / Glioblastoma / Jumonji Domain-Containing Histone Demethylases Type of study: Clinical_trials Limits: Animals / Humans Language: En Journal: Cell Death Dis Year: 2021 Type: Article