Dual role of proliferating cell nuclear antigen monoubiquitination in facilitating Fanconi anemia-mediated interstrand crosslink repair

Author:

Shah Ronak1,Aslam Muhammad Assad12ORCID,Spanjaard Aldo1ORCID,de Groot Daniel1,Zürcher Lisa M1ORCID,Altelaar Maarten34,Hoekman Liesbeth3ORCID,Pritchard Colin E J5,Pilzecker Bas1ORCID,van den Berk Paul C M1ORCID,Jacobs Heinz1ORCID

Affiliation:

1. Department of Tumor Biology and Immunology, The Netherlands Cancer Institute , Plesmanlaan 121, 1066 CX Amsterdam , The Netherlands

2. Department/Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University , Bosan Road, 60800 Multan , Pakistan

3. Proteomics Facility, The Netherlands Cancer Institute , Plesmanlaan 121, 1066CX Amsterdam , The Netherlands

4. Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University and Netherlands Proteomics Centre , Utrecht, Padualaan 8, 3584 CH Utrecht , The Netherlands

5. Mouse Clinic for Cancer and Aging Transgenic Facility, The Netherlands Cancer Institute , Plesmanlaan 121, 1066 CX Amsterdam , The Netherlands

Abstract

Abstract The Fanconi anemia (FA) repair pathway governs repair of highly genotoxic DNA interstrand crosslinks (ICLs) and relies on translesion synthesis (TLS). TLS is facilitated by REV1 or site-specific monoubiquitination of proliferating cell nuclear antigen (PCNA) (PCNA-Ub) at lysine 164 (K164). A PcnaK164R/K164R but not Rev1−/− mutation renders mammals hypersensitive to ICLs. Besides the FA pathway, alternative pathways have been associated with ICL repair (1, 2), though the decision making between those remains elusive. To study the dependence and relevance of PCNA-Ub in FA repair, we intercrossed PcnaK164R/+; Fancg−/+ mice. A combined mutation (PcnaK164R/K164R; Fancg−/−) was found embryonically lethal. RNA-seq of primary double-mutant (DM) mouse embryonic fibroblasts (MEFs) revealed elevated levels of replication stress-induced checkpoints. To exclude stress-induced confounders, we utilized a Trp53 knock-down to obtain a model to study ICL repair in depth. Regarding ICL-induced cell toxicity, cell cycle arrest, and replication fork progression, single-mutant and DM MEFs were found equally sensitive, establishing PCNA-Ub to be critical for FA-ICL repair. Immunoprecipitation and spectrometry-based analysis revealed an unknown role of PCNA-Ub in excluding mismatch recognition complex MSH2/MSH6 from being recruited to ICLs. In conclusion, our results uncovered a dual function of PCNA-Ub in ICL repair, i.e. exclude MSH2/MSH6 recruitment to channel the ICL toward canonical FA repair, in addition to its established role in coordinating TLS opposite the unhooked ICL.

Funder

The Dutch Cancer Society

Dutch Ministry of Health, Welfare and Sport

Publisher

Oxford University Press (OUP)

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3