Publication

c-Jun-N-terminal phosphorylation regulates DNMT1 expression and genome wide methylation in gliomas

Journal Paper/Review - Jan 24, 2017

Units
PubMed
Doi

Citation
Heiland D, Nelander S, Kling T, Weyerbrock A, Kling E, Masilamani A, Dai F, Claus R, Ferrarese R, Carro M. c-Jun-N-terminal phosphorylation regulates DNMT1 expression and genome wide methylation in gliomas. Oncotarget 2017; 8:6940-6954.
Type
Journal Paper/Review (English)
Journal
Oncotarget 2017; 8
Publication Date
Jan 24, 2017
Issn Electronic
1949-2553
Pages
6940-6954
Brief description/objective

High-grade gliomas (HGG) are the most common brain tumors, with an average survival time of 14 months. A glioma-CpG island methylator phenotype (G-CIMP), associated with better clinical outcome, has been described in low and high-grade gliomas. Mutation of IDH1 is known to drive the G-CIMP status. In some cases, however, the hypermethylation phenotype is independent of IDH1 mutation, suggesting the involvement of other mechanisms. Here, we demonstrate that DNMT1 expression is higher in low-grade gliomas compared to glioblastomas and correlates with phosphorylated c-Jun. We show that phospho-c-Jun binds to the DNMT1 promoter and causes DNA hypermethylation. Phospho-c-Jun activation by Anisomycin treatment in primary glioblastoma-derived cells attenuates the aggressive features of mesenchymal glioblastomas and leads to promoter methylation and downregulation of key mesenchymal genes (CD44, MMP9 and CHI3L1). Our findings suggest that phospho-c-Jun activates an important regulatory mechanism to control DNMT1 expression and regulate global DNA methylation in Glioblastoma.