Your browser doesn't support javascript.
loading
HLTF Promotes Fork Reversal, Limiting Replication Stress Resistance and Preventing Multiple Mechanisms of Unrestrained DNA Synthesis.
Bai, Gongshi; Kermi, Chames; Stoy, Henriette; Schiltz, Carl J; Bacal, Julien; Zaino, Angela M; Hadden, M Kyle; Eichman, Brandt F; Lopes, Massimo; Cimprich, Karlene A.
Affiliation
  • Bai G; Department of Chemical and Systems Biology, Stanford University School of Medicine, 318 Campus Drive, Stanford, CA 94305-5441, USA.
  • Kermi C; Department of Chemical and Systems Biology, Stanford University School of Medicine, 318 Campus Drive, Stanford, CA 94305-5441, USA.
  • Stoy H; Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland.
  • Schiltz CJ; Department of Biological Sciences and Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA.
  • Bacal J; Department of Chemical and Systems Biology, Stanford University School of Medicine, 318 Campus Drive, Stanford, CA 94305-5441, USA.
  • Zaino AM; Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06029-3092, USA.
  • Hadden MK; Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06029-3092, USA.
  • Eichman BF; Department of Biological Sciences and Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA.
  • Lopes M; Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland.
  • Cimprich KA; Department of Chemical and Systems Biology, Stanford University School of Medicine, 318 Campus Drive, Stanford, CA 94305-5441, USA. Electronic address: cimprich@stanford.edu.
Mol Cell ; 78(6): 1237-1251.e7, 2020 06 18.
Article in En | MEDLINE | ID: mdl-32442397
ABSTRACT
DNA replication stress can stall replication forks, leading to genome instability. DNA damage tolerance pathways assist fork progression, promoting replication fork reversal, translesion DNA synthesis (TLS), and repriming. In the absence of the fork remodeler HLTF, forks fail to slow following replication stress, but underlying mechanisms and cellular consequences remain elusive. Here, we demonstrate that HLTF-deficient cells fail to undergo fork reversal in vivo and rely on the primase-polymerase PRIMPOL for repriming, unrestrained replication, and S phase progression upon limiting nucleotide levels. By contrast, in an HLTF-HIRAN mutant, unrestrained replication relies on the TLS protein REV1. Importantly, HLTF-deficient cells also exhibit reduced double-strand break (DSB) formation and increased survival upon replication stress. Our findings suggest that HLTF promotes fork remodeling, preventing other mechanisms of replication stress tolerance in cancer cells. This remarkable plasticity of the replication fork may determine the outcome of replication stress in terms of genome integrity, tumorigenesis, and response to chemotherapy.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Transcription Factors / DNA / DNA-Binding Proteins / DNA Replication Limits: Humans Language: En Journal: Mol Cell Journal subject: BIOLOGIA MOLECULAR Year: 2020 Document type: Article Affiliation country: Estados Unidos

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Transcription Factors / DNA / DNA-Binding Proteins / DNA Replication Limits: Humans Language: En Journal: Mol Cell Journal subject: BIOLOGIA MOLECULAR Year: 2020 Document type: Article Affiliation country: Estados Unidos