Abstract

BackgroundProper repair and restart of stressed replication forks requires intact homologous recombination (HR). HR at stressed replication forks can be initiated by the 5′ endonuclease EEPD1, which cleaves the stalled replication fork. Inherited or acquired defects in HR, such as mutations in breast cancer susceptibility protein-1 (BRCA1) or BRCA2, predispose to cancer, including breast and ovarian cancers. In order for these HR-deficient tumor cells to proliferate, they become addicted to a bypass replication fork repair pathway mediated by radiation repair protein 52 (RAD52). Depleting RAD52 can cause synthetic lethality in BRCA1/2 mutant cancers by an unknown molecular mechanism.MethodsWe hypothesized that cleavage of stressed replication forks by EEPD1 generates a fork repair intermediate that is toxic when HR-deficient cells cannot complete repair with the RAD52 bypass pathway. To test this hypothesis, we applied cell survival assays, immunofluorescence staining, DNA fiber and western blot analyses to look at the correlation between cell survival and genome integrity in control, EEPD1, RAD52 and EEPD1/RAD52 co-depletion BRCA1-deficient breast cancer cells.ResultsOur data show that depletion of EEPD1 suppresses synthetic lethality, genome instability, mitotic catastrophe, and hypersensitivity to stress of replication of RAD52-depleted, BRCA1 mutant breast cancer cells. Without HR and the RAD52-dependent backup pathway, the BRCA1 mutant cancer cells depleted of EEPD1 skew to the alternative non-homologous end-joining DNA repair pathway for survival.ConclusionThis study indicates that the mechanism of synthetic lethality in RAD52-depleted BRCA1 mutant cancer cells depends on the endonuclease EEPD1. The data imply that EEPD1 cleavage of stressed replication forks may result in a toxic intermediate when replication fork repair cannot be completed.

Highlights

  • Proper repair and restart of stressed replication forks requires intact homologous recombination (HR)

  • HR is mediated by a litany of components that are regulated by breast cancer susceptibility protein-1 (BRCA1), which promotes the initial step in HR, 5′ end Hromas et al Breast Cancer Research (2017) 19:122 resection to create 3′ single-stranded (SS) DNA

  • Depletion of Endonuclease/exonuclease/ phosphatase family domain-containing-1 (EEPD1) promotes survival in BRCA1-mutated and Radiation repair protein 52 (RAD52)-depleted breast cancer cells MDA-MB-436 BRCA1-/- breast cancer cells were depleted of EEPD1 and/or RAD52 using small interfering RNA (siRNA)

Read more

Summary

Introduction

Proper repair and restart of stressed replication forks requires intact homologous recombination (HR). Inherited or acquired defects in HR, such as mutations in breast cancer susceptibility protein-1 (BRCA1) or BRCA2, predispose to cancer, including breast and ovarian cancers. In order for these HR-deficient tumor cells to proliferate, they become addicted to a bypass replication fork repair pathway mediated by radiation repair protein 52 (RAD52). Radiation repair protein 51 (RAD51)-dependent homologous recombination (HR) is the canonical repair and restart pathway for stalled replications forks [6,7,8,9]. HR is mediated by a litany of components that are regulated by breast cancer susceptibility protein-1 (BRCA1), which promotes the initial step in HR, 5′ end Hromas et al Breast Cancer Research (2017) 19:122 resection to create 3′ single-stranded (SS) DNA. After the invading strand re-initiates DNA replication, HR intermediates such as Holliday junctions are resolved by Holliday junction 5′ flap endonuclease (GEN1) or MUS81 structure-specific endonuclease subunit (MUS81), with SLX4 structure-specific endonuclease subunit (SLX4) serving as a scaffold [11,12,13]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call