首页 | 本学科首页   官方微博 | 高级检索  
   检索      


USP19 Deubiquitinating Enzyme Supports Cell Proliferation by Stabilizing KPC1, a Ubiquitin Ligase for p27Kip1
Authors:Yu Lu  Olasunkanmi A J Adegoke  Alain Nepveu  Keiichi I Nakayama  Nathalie Bedard  Dongmei Cheng  Junmin Peng  Simon S Wing
Institution:Polypeptide Laboratory, Division of Endocrinology and Metabolism,1. Molecular Oncology Group, Department of Medicine, McGill University and McGill University Health Centre, Montreal, Quebec, Canada H3A 2B2,2. Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan,3. Department of Human Genetics, Center for Neurodegenerative Disease, Emory University, Atlanta, Georgia 303224.
Abstract:p27Kip1 is a cyclin-dependent kinase inhibitor that regulates the G1/S transition. Increased degradation of p27Kip1 is associated with cellular transformation. Previous work demonstrated that the ubiquitin ligases KPC1/KPC2 and SCFSkp2 ubiquitinate p27Kip1 in G1 and early S, respectively. The regulation of these ligases remains unclear. We report here that the USP19 deubiquitinating enzyme interacts with and stabilizes KPC1, thereby modulating p27Kip1 levels and cell proliferation. Cells depleted of USP19 by RNA interference exhibited an inhibition of cell proliferation, progressing more slowly from G0/G1 to S phase, and accumulated p27Kip1. This increase in p27Kip1 was associated with normal levels of Skp2 but reduced levels of KPC1. The overexpression of KPC1 or the use of p27−/− cells inhibited significantly the growth defect observed upon USP19 depletion. KPC1 was ubiquitinated in vivo and stabilized by proteasome inhibitors and by overexpression of USP19, and it also coimmunoprecipitated with USP19. Our results identify USP19 as the first deubiquitinating enzyme that regulates the stability of a cyclin-dependent kinase inhibitor and demonstrate that progression through G1 to S phase is, like the metaphase-anaphase transition, controlled in a hierarchical, multilayered fashion.The ubiquitin proteasome pathway plays essential roles in regulating the cell cycle. The best-defined functions of this pathway in cell cycle regulation are those mediated by the multisubunit ubiquitin protein ligases SKP1-CUL1-F-box (SCF) and the anaphase-promoting complex/cyclosome (APC) (reviewed in reference 25). The functions of the APC in the cell cycle are predominant at the mitosis-anaphase transition, while the activities of SCF-type ubiquitin protein ligase complexes are involved at various steps of the cycle (reviewed in references 21 and 25). One of the best-defined functions of the SCF is mediated by SCFskp2, which plays a vital role in regulating the G1-S transition by ubiquitinating the cyclin-dependent kinase inhibitor p27Kip1, thereby targeting it for degradation by the proteasome (3, 31, 32).The central role of p27Kip1 in restricting cell proliferation is demonstrated by the fact that mice lacking the p27Kip1 gene manifest increased body and organ weights and develop pituitary adenomas (6, 13, 18). In addition, the results of clinical studies suggest that low p27Kip1 levels are associated with increased aggressivity of tumors (1, 28). Unlike the case with the p53 or Rb tumor suppressors, mutation or deletion of p27Kip1 in tumors is rare. Rather, its deregulation in human tumors is due mainly to reduced protein levels, mediated in large part by increased proteolysis (reviewed in reference 21). In support of this, the low p27Kip1 levels seen in tumors are associated with increased levels of Skp2, the substrate recognition subunit of the SCFskp2 ligase. The loss of Skp2 in mice results in p27Kip1 accumulation, and cells from Skp2/ animals contain enlarged nuclei with polyploidy and multiple centrosomes. They also show a reduced growth rate and increased apoptosis (19). Many of the cellular phenotypes observed in Skp2/ mice disappear in Skp2/ p27/ double-mutant mice (14, 20). Thus, the oncogenic nature of Skp2 is largely due to its ability to mediate p27Kip1 degradation.In spite of the clear role of SCFskp2 in mediating the ubiquitination and degradation of p27Kip1, the downregulation of this cyclin-dependent kinase inhibitor proceeds normally in lymphocytes isolated from Skp2−/− mice (8, 21). In addition, in the normal cell cycle, p27Kip1 is also degraded in G1, before the expression of Skp2, which occurs in early S phase (8, 9, 36). Also, p27Kip1 is exported from the nucleus to the cytoplasm in G1, whereas Skp2 is localized in the nucleus (9, 26). These observations suggest the existence of another pathway for the degradation of p27Kip1. Indeed, KPC (Kip1 ubiquitination-promoting complex) was subsequently identified as a novel cytoplasmic ligase complex that interacts with and ubiquitinates p27Kip1 (10). KPC consists of two subunits: KPC1, a 140-kDa RING-finger domain-containing protein, and KPC2, a 50-kDa protein containing a ubiquitin-like domain and two ubiquitin-associated domains.Although the role of ubiquitin protein ligases in the cell cycle has received considerable attention, fewer data are available regarding the roles of deubiquitinating enzymes in the cell cycle (22). We recently described USP19 as a deubiquitinating enzyme that is induced in skeletal muscle atrophying in response to numerous catabolic stimuli. To study its function, we used RNA interference to explore the consequences of depletion of this enzyme in cultured muscle cells. Our early studies indicated that the loss of USP19 interfered with the growth of L6 myoblasts. We have observed similar effects in FR3T3 fibroblasts and have explored the underlying mechanisms of this growth defect.
Keywords:
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号