Nature Communications (Jun 2024)

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

  • Samuel D. Chauvin,
  • Shoichiro Ando,
  • Joe A. Holley,
  • Atsushi Sugie,
  • Fang R. Zhao,
  • Subhajit Poddar,
  • Rei Kato,
  • Cathrine A. Miner,
  • Yohei Nitta,
  • Siddharth R. Krishnamurthy,
  • Rie Saito,
  • Yue Ning,
  • Yuya Hatano,
  • Sho Kitahara,
  • Shin Koide,
  • W. Alexander Stinson,
  • Jiayuan Fu,
  • Nehalee Surve,
  • Lindsay Kumble,
  • Wei Qian,
  • Oleksiy Polishchuk,
  • Prabhakar S. Andhey,
  • Cindy Chiang,
  • Guanqun Liu,
  • Ludovic Colombeau,
  • Raphaël Rodriguez,
  • Nicolas Manel,
  • Akiyoshi Kakita,
  • Maxim N. Artyomov,
  • David C. Schultz,
  • P. Toby Coates,
  • Elisha D. O. Roberson,
  • Yasmine Belkaid,
  • Roger A. Greenberg,
  • Sara Cherry,
  • Michaela U. Gack,
  • Tristan Hardy,
  • Osamu Onodera,
  • Taisuke Kato,
  • Jonathan J. Miner

DOI
https://doi.org/10.1038/s41467-024-49066-7
Journal volume & issue
Vol. 15, no. 1
pp. 1 – 23

Abstract

Read online

Abstract Age-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.