Rewired m6A epitranscriptomic networks link mutant p53 to neoplastic transformation

An Xu,Mo Liu, Mo-Fan Huang,Yang Zhang, Ruifeng Hu,Julian A. Gingold, Ying Liu,Dandan Zhu, Chian-Shiu Chien,Wei-Chen Wang,Zian Liao, Fei Yuan,Chih-Wei Hsu, Jian Tu,Yao Yu, Taylor Rosen, Feng Xiong,Peilin Jia, Yi-Ping Yang,Danielle A. Bazer,Ya-Wen Chen, Wenbo Li,Chad D. Huff, Jay-Jiguang Zhu,Francesca Aguilo, Shih-Hwa Chiou,Nathan C. Boles, Chien-Chen Lai,Mien-Chie Hung, Zhongming Zhao,Eric L. Van Nostrand, Ruiying Zhao,Dung-Fang Lee

NATURE COMMUNICATIONS(2023)

引用 0|浏览12
暂无评分
摘要
N6-methyladenosine (m(6)A), one of themost prevalent mRNA modifications in eukaryotes, plays a critical role in modulating both biological and pathological processes. However, it is unknown whether mutant p53 neomorphic oncogenic functions exploit dysregulation of m(6)A epitranscriptomic networks. Here, we investigate Li-Fraumeni syndrome (LFS)-associated neoplastic transformation driven by mutant p53 in iPSC-derived astrocytes, the cell-of-origin of gliomas. We find that mutant p53 but not wild-type (WT) p53 physically interacts with SVIL to recruit the H3K4me3 methyltransferase MLL1 to activate the expression of m(6)A reader YTHDF2, culminating in an oncogenic phenotype. Aberrant YTHDF2 upregulation markedly hampers expression of multiple m(6)A-marked tumor-suppressing transcripts, including CDKN2B and SPOCK2, and induces oncogenic reprogramming. Mutant p53 neoplastic behaviors are significantly impaired by genetic depletion of YTHDF2 or by pharmacological inhibition using MLL1 complex inhibitors. Our study reveals howmutant p53 hijacks epigenetic and epitranscriptomicmachinery to initiate gliomagenesis and suggests potential treatment strategies for LFS gliomas.
更多
查看译文
关键词
CNS cancer,Induced pluripotent stem cells,Tumour-suppressor proteins,Science,Humanities and Social Sciences,multidisciplinary
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要