The long-term goal of this research program is to understand how human cells respond to replication stress, the slowing or stalling of DNA replication induced by barriers to replication fork progression. Fork stalling barriers can be induced environmentally by alkylating agents, crosslinking agents and aldehydes, or they can arise from endogenous DNA damage, secondary DNA structures or interference with transcription. Prolonged replication fork stalling can lead to DNA break formation and a failure to complete DNA replication. This is turn can result in genome instability and mutagenesis, properties that drive cancers and affect their sensitivity to chemotherapy. Fortunately, cells can respond to replication stress in multiple ways, depending on the barrier encountered. Two crucial components of the replication stress response involve fork slowing and replication fork reversal, a process that involves reannealing of the unwound template DNA and annealing of nascent DNA strands. Fork reversal is thought to promote DNA repair, stabilize the replication fork and facilitate certain forms of fork restart. It can also allow for the error-free replication of damaged DNA by providing the undamaged sister chromatid as a template for DNA replication. When fork reversal occurs and how it is regulated in the cell are poorly understood. In the previous funding period, we made the surprising finding that the DNA translocase and fork reversal enzyme, HLTF, is needed to slow replication forks during replication stress. This fork slowing requires HLTF's fork reversal activity. HLTF-deficient cells also proliferate better under replication stress conditions. This combination of unusual phenotypes is unique to HLTF among fork remodelers. The objective of this application is to characterize this stress-resistant, potentially error-prone replication mode at the molecular and cellular level, thereby addressing fundamental questions about the balance of replication fork progression and fork reversal during replication stress. In the first aim, we will explore how HLTF processes replication forks to slow their progression and induce DNA breaks. In the second aim, we will determine the mechanisms and mutagenic consequences of stress-resistant DNA replication and unrestrained fork progression. Finally, in the third aim, we will probe the mechanisms of cell survival under replication stress, searching for targets that contribute to stress resistance in HLTF-deficient cells. These experiments will capitalize on cutting-edge genomic and proteomic approaches to solve fundamental problems regarding replication fork reversal and replication stress resistance. !

Public Health Relevance

Accurate DNA replication through environmental and endogenous challenges is essential to prevent cancer, premature aging and other diseases. HLTF is silenced in a number of colorectal and stomach cancers. Understanding how HLTF loss affects cells and how HLTF mechanistically restricts replication and mutagenesis could ultimately lead to new therapeutic approaches to treat these and other cancers exhibiting resistance to replication stress. !

Agency
National Institute of Health (NIH)
Institute
National Institute of Environmental Health Sciences (NIEHS)
Type
Research Project (R01)
Project #
5R01ES016486-17
Application #
9656122
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Reinlib, Leslie J
Project Start
2002-05-01
Project End
2023-03-31
Budget Start
2019-04-01
Budget End
2020-03-31
Support Year
17
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Stanford University
Department
Pharmacology
Type
Schools of Medicine
DUNS #
009214214
City
Stanford
State
CA
Country
United States
Zip Code
94305
Saldivar, Joshua C; Cimprich, Karlene A (2018) A new mitotic activity comes into focus. Science 359:30-31
Saldivar, Joshua C; Hamperl, Stephan; Bocek, Michael J et al. (2018) An intrinsic S/G2 checkpoint enforced by ATR. Science 361:806-810
Saldivar, Joshua C; Cortez, David; Cimprich, Karlene A (2017) The essential kinase ATR: ensuring faithful duplication of a challenging genome. Nat Rev Mol Cell Biol 18:622-636
Hess, Gaelen T; Frésard, Laure; Han, Kyuho et al. (2016) Directed evolution using dCas9-targeted somatic hypermutation in mammalian cells. Nat Methods 13:1036-1042
Kile, Andrew C; Chavez, Diana A; Bacal, Julien et al. (2015) HLTF's Ancient HIRAN Domain Binds 3' DNA Ends to Drive Replication Fork Reversal. Mol Cell 58:1090-100
Slaats, Gisela G; Saldivar, Joshua C; Bacal, Julien et al. (2015) DNA replication stress underlies renal phenotypes in CEP290-associated Joubert syndrome. J Clin Invest 125:3657-66
Zeman, Michelle K; Lin, Jia-Ren; Freire, Raimundo et al. (2014) DNA damage-specific deubiquitination regulates Rad18 functions to suppress mutagenesis. J Cell Biol 206:183-97
Zeman, Michelle K; Cimprich, Karlene A (2014) Causes and consequences of replication stress. Nat Cell Biol 16:2-9
Duursma, Anja M; Driscoll, Robert; Elias, Josh E et al. (2013) A role for the MRN complex in ATR activation via TOPBP1 recruitment. Mol Cell 50:116-22
Machado, Luciana E F; Pustovalova, Yulia; Kile, Andrew C et al. (2013) PHD domain from human SHPRH. J Biomol NMR 56:393-9

Showing the most recent 10 out of 23 publications