Abstract

End resection of DNA double-strand breaks (DSBs) to form 3′ single-strand DNA (ssDNA) is critical to initiate the homologous recombination (HR) pathway of DSB repair. HR pathway is strictly limited in the G1-phase cells because of lack of homologous DNA as the templates. Exonuclease 1 (EXO1) is the key molecule responsible for 3′ ssDNA formation of DSB end resection. We revealed that EXO1 is inactivated in G1-phase cells via ubiquitination-mediated degradation, resulting from an elevated expression level of RING-box protein 1 (RBX1) in G1 phase. The increased RBX1 significantly prompted the neddylation of Cullin1 and contributed to the G1 phase-specific degradation of EXO1. Knockdown of RBX1 remarkedly attenuated the degradation of EXO1 and increased the end resection and HR activity in γ-irradiated G1-phase cells, as demonstrated by the increased formation of RPA32, BrdU, and RAD51 foci. And EXO1 depletion mitigated DNA repair defects due to RBX1 reduction. Moreover, increased autophosphorylation of DNA-PKcs at S2056 was found to be responsible for the higher expression level of the RBX1 in the G1 phase. Inactivation of DNA-PKcs decreased RBX1 expression, and simultaneously increased EXO1 expression and DSB end resection in G1-phase cells. This study demonstrates a new mechanism for restraining the HR pathway of DNA DSB repair in G1 phase via RBX1-prompted inactivation of EXO1.

Highlights

  • DNA double-strand breaks (DSBs) are among the most lethal types of DNA damage

  • To illustrate how cells would benefit from Exonuclease 1 (EXO1) degradation after IR exposure, we further investigated the effects of RING-box protein 1 (RBX1) on end resection and homologous recombination (HR) repair in

  • EXO1 is a critical component in the end resection of the HR pathway of DNA DSB repair

Read more

Summary

Introduction

DNA double-strand breaks (DSBs) are among the most lethal types of DNA damage. Failure to properly repair DSBs leads to chromosomal aberrations, an overall increase in genomic instability and cell death [1,2,3,4,5]. In G1 phase, NHEJ is preferably adapted to repair DSBs, while HR activity is tightly restrained due to the lack of necessary templates for homologous DNA. It is still an important question how to guarantee the proper DSB repair pathway choice during the cell cycle. Despite its critical role in inducing the overall DNA damage response, it is important to understand whether and how EXO1 nuclease plays a role in restraining HR activity in G1-phase cells to guarantee the proper choice of the DSB repair pathway. Fluorescence-conjugated anti-mouse immunoglobulin G (IgG) and anti-rabbit immunoglobulin G (IgG) were purchased from Molecular Probes (Alexa Fluor 488 and 568)

Material and methods
Results
Discussion
Findings
Compliance with ethical standards
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