Abstract

Proper cell division relies on the coordinated regulation between a structural component, the mitotic spindle, and a regulatory component, anaphase-promoting complex/cyclosome (APC/C). Hematopoietic PBX-interacting protein (HPIP) is a microtubule-associated protein that plays a pivotal role in cell proliferation, cell migration, and tumor metastasis. Here, using HEK293T and HeLa cells, along with immunoprecipitation and immunoblotting, live-cell imaging, and protein-stability assays, we report that HPIP expression oscillates throughout the cell cycle and that its depletion delays cell division. We noted that by utilizing its D box and IR domain, HPIP plays a dual role both as a substrate and inhibitor, respectively, of the APC/C complex. We observed that HPIP enhances the G2/M transition of the cell cycle by transiently stabilizing cyclin B1 by preventing APC/C-Cdc20-mediated degradation, thereby ensuring timely mitotic entry. We also uncovered that HPIP associates with the mitotic spindle and that its depletion leads to the formation of multiple mitotic spindles and chromosomal abnormalities, results in defects in cytokinesis, and delays mitotic exit. Our findings uncover HPIP as both a substrate and an inhibitor of APC/C-Cdc20 that maintains the temporal stability of cyclin B1 during the G2/M transition and thereby controls mitosis and cell division.

Highlights

  • Proper cell division relies on the coordinated regulation between a structural component, the mitotic spindle, and a regulatory component, anaphase-promoting complex/cyclosome (APC/C)

  • These results indicated that Hematopoietic PBX-interacting protein (HPIP) expression is required for normal cell division, and HPIP may act as a G2/M transition regulator

  • Our study demonstrates that HPIP is a critical regulator of G2/M transition by regulating temporal stability of cyclin B1 via inhibition of APC/C–Cdc20 activity

Read more

Summary

Role of HPIP in cell cycle regulation

Cell cycle [21, 22]. The precise mechanism by which HPIP regulates cell cycle progression remains elusive. We demonstrated that HPIP protein levels oscillate during various stages of cell cycle and HPIP is degraded during mitosis by APC/C–Cdc but not by APC/C–Cdh. This study demonstrates that HPIP associates with mitotic spindle, and its depletion leads to the formation of multiple spindle poles, chromosomal abnormality, and delayed mitotic exit. These studies suggest a dual role for HPIP as substrate and an inhibitor of APC/ C–Cdc during cell cycle progression

Loss of HPIP expression delays cell division
HPIP protein level oscillates during cell cycle
HPIP has spindle check point function
Loss of HPIP expression leads to defects in cytokinesis
Discussion
Role of HPIP in mitotic checkpoint function
Experimental procedures
Cell synchronization and cycle analysis
Live cell imaging for cell cycle progression
Immunoprecipitation and Western blotting
Ubiquitination assays
Protein stability and chasing experiments
Confocal microscopy
Metaphase spread preparation
Statistical analysis
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