Abstract

FBXO31 was originally identified as a putative tumor suppressor gene in breast, ovarian, hepatocellular, and prostate cancers. By screening a set of cell cycle-regulated proteins as potential FBXO31 interaction partners, we have now identified Cdt1 as a novel substrate. Cdt1 DNA replication licensing factor is part of the pre-replication complex and essential for the maintenance of genomic integrity. We show that FBXO31 specifically interacts with Cdt1 and regulates its abundance by ubiquitylation leading to subsequent degradation. We also show that Cdt1 regulation by FBXO31 is limited to the G2 phase of the cell cycle and is independent of the pathways previously described for Cdt1 proteolysis in S and G2 phase. FBXO31 targeting of Cdt1 is mediated through the N terminus of Cdt1, a region previously shown to be responsible for its cell cycle regulation. Finally, we show that Cdt1 stabilization due to FBXO31 depletion results in re-replication. Our data present an additional pathway that contributes to the FBXO31 function as a tumor suppressor.

Highlights

  • SCF E3 ligases regulate degradation of proteins involved in many processes including cell cycle progression and DNA repair

  • We show that Cdt1 regulation by FBXO31 is limited to the G2 phase of the cell cycle and is independent of the pathways previously described for Cdt1 proteolysis in S and G2 phase

  • Protein extracts from HEK293T cells transiently expressing Myc-tagged Cdt1 were subjected to immunoprecipitation with either anti-Myc antibody or IgG control and Western blotted with an anti-FBXO31 antibody

Read more

Summary

Background

SCF E3 ligases regulate degradation of proteins involved in many processes including cell cycle progression and DNA repair. By screening a set of cell cycle-regulated proteins as potential FBXO31 interaction partners, we have identified Cdt as a novel substrate. The activity and protein levels of Cdt are tightly controlled in S and G2 phases of the cell cycle through ubiquitylation-dependent proteolysis by SCF-Skp ubiquitin ligase and the proliferating cell nuclear antigen-dependent Cul4-DDB1 and Cdt ubiquitin ligases as well as the inhibitory geminin binding to prevent rereplication and genomic instability (18, 19). Cdt regulation by FBXO31 is limited to the G2 and the early mitotic phase of the cell cycle and is independent of the pathways previously described for Cdt proteolysis in S and G2 phase. Our data present a novel pathway that regulates Cdt in the G2 phase of the cell cycle and is likely to contribute to FBXO31 tumor suppressor activity

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.