Inherited C-terminal TREX1 variants disrupt homology-directed repair to cause senescence and DNA damage phenotypes in Drosophila, mice, and humans

Author:

Chauvin Samuel D.ORCID,Ando Shoichiro,Holley Joe A.,Sugie AtsushiORCID,Zhao Fang R.ORCID,Poddar Subhajit,Kato Rei,Miner Cathrine A.,Nitta YoheiORCID,Krishnamurthy Siddharth R.,Saito RieORCID,Ning Yue,Hatano Yuya,Kitahara Sho,Koide ShinORCID,Stinson W. Alexander,Fu Jiayuan,Surve Nehalee,Kumble Lindsay,Qian Wei,Polishchuk Oleksiy,Andhey Prabhakar S.,Chiang Cindy,Liu GuanqunORCID,Colombeau Ludovic,Rodriguez RaphaëlORCID,Manel NicolasORCID,Kakita Akiyoshi,Artyomov Maxim N.ORCID,Schultz David C.ORCID,Coates P. Toby,Roberson Elisha D. O.,Belkaid Yasmine,Greenberg Roger A.ORCID,Cherry SaraORCID,Gack Michaela U.ORCID,Hardy Tristan,Onodera OsamuORCID,Kato TaisukeORCID,Miner Jonathan J.ORCID

Abstract

AbstractAge-related microangiopathy, also known as small vessel disease (SVD), causes damage to the brain, retina, liver, and kidney. Based on the DNA damage theory of aging, we reasoned that genomic instability may underlie an SVD caused by dominant C-terminal variants in TREX1, the most abundant 3′−5′ DNA exonuclease in mammals. C-terminal TREX1 variants cause an adult-onset SVD known as retinal vasculopathy with cerebral leukoencephalopathy (RVCL or RVCL-S). In RVCL, an aberrant, C-terminally truncated TREX1 mislocalizes to the nucleus due to deletion of its ER-anchoring domain. Since RVCL pathology mimics that of radiation injury, we reasoned that nuclear TREX1 would cause DNA damage. Here, we show that RVCL-associated TREX1 variants trigger DNA damage in humans, mice, and Drosophila, and that cells expressing RVCL mutant TREX1 are more vulnerable to DNA damage induced by chemotherapy and cytokines that up-regulate TREX1, leading to depletion of TREX1-high cells in RVCL mice. RVCL-associated TREX1 mutants inhibit homology-directed repair (HDR), causing DNA deletions and vulnerablility to PARP inhibitors. In women with RVCL, we observe early-onset breast cancer, similar to patients with BRCA1/2 variants. Our results provide a mechanistic basis linking aberrant TREX1 activity to the DNA damage theory of aging, premature senescence, and microvascular disease.

Funder

U.S. Department of Health & Human Services | NIH | National Institute of Arthritis and Musculoskeletal and Skin Diseases

U.S. Department of Health & Human Services | NIH | National Institute of Allergy and Infectious Diseases

U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke

Rheumatology Research Foundation

The Clayco Foundation Penn Colton Center for Autoimmunity Penn RVCL Sisters Fund

U.S. Department of Health & Human Services | National Institutes of Health

Japan Agency for Medical Research and Development

MEXT | Japan Society for the Promotion of Science

Osaka Medical Research Foundation for Intractable Diseases

Publisher

Springer Science and Business Media LLC

Cited by 1 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

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