Advanced Search

Submit Manuscript

Volume 28, No 7, Jul 2018

ISSN: 1001-0602 
EISSN: 1748-7838 2018 
impact factor 17.848* 
(Clarivate Analytics, 2019)

Volume 28 Issue 7, July 2018: 756-770   |  Open Access

ORIGINAL ARTICLES

Destabilization of linker histone H1.2 is essential for ATM activation and DNA damage repair

Zhiming Li 1 , Yinglu Li 1, Ming Tang 1, Bin Peng 2, Xiaopeng Lu 2, Qiaoyan Yang 1, Qian Zhu 2 , Tianyun Hou 1,2 , Meiting Li 1 , Chaohua Liu 1 ,Lina Wang 1 , Xingzhi Xu 2 , Ying Zhao 1 , Haiying Wang 1 , Yang Yang 1 and Wei-Guo Zhu 1,2

1Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China and 2 Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen 518060, China
Correspondence: Correspondence: Wei-Guo Zhu (zhuweiguo@szu.edu.cn)These authors contributed equally: Zhiming Li, Yinglu Li

Linker histone H1 is a master regulator of higher order chromatin structure, but its involvement in the DNA damage response and repair is unclear. Here, we report that linker histone H1.2 is an essential regulator of ataxia telangiectasia mutated (ATM) activation. We show that H1.2 protects chromatin from aberrant ATM activation through direct interaction with the ATM HEAT repeat domain and inhibition of MRE11-RAD50-NBS1 (MRN) complex-dependent ATM recruitment. Upon DNA damage, H1.2 undergoes rapid PARP1-dependent chromatin dissociation through poly-ADP-ribosylation (PARylation) of its C terminus and further proteasomal degradation. Inhibition of H1.2 displacement by PARP1 depletion or an H1.2 PARylation-dead mutation compromises ATM activation and DNA damage repair, thus leading to impaired cell survival. Taken together, our findings suggest that linker histone H1.2 functions as a physiological barrier for ATM to target the chromatin, and PARylation-mediated active H1.2 turnover is required for robust ATM activation and DNA damage repair.


https://doi.org/10.1038/s41422-018-0048-0

FULL TEXT | PDF

Browse 1315