Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1865(1): 105-116, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28939057

ABSTRACT

The Cdc28 protein kinase subunits, Cks1 and Cks2, play dual roles in Cdk-substrate specificity and Cdk-independent protein degradation, in concert with the E3 ubiquitin ligase complexes SCFSkp2 and APCCdc20. Notable targets controlled by Cks include p27 and Cyclin A. Here, we demonstrate that Cks1 and Cks2 proteins interact with both the MllN and MllC subunits of Mll1 (Mixed-lineage leukaemia 1), and together, the Cks proteins define Mll1 levels throughout the cell cycle. Overexpression of CKS1B and CKS2 is observed in multiple human cancers, including various MLL-rearranged (MLLr) AML subtypes. To explore the importance of MLL-Fusion Protein regulation by CKS1/2, we used small molecule inhibitors (MLN4924 and C1) to modulate their protein degradation functions. These inhibitors specifically reduced the proliferation of MLLr cell lines compared to primary controls. Altogether, this study uncovers a novel regulatory pathway for MLL1, which may open a new therapeutic approach to MLLr leukaemia.


Subject(s)
CDC2-CDC28 Kinases/physiology , Carrier Proteins/physiology , Cell Cycle Proteins/physiology , Histone-Lysine N-Methyltransferase/genetics , Leukemia/genetics , Leukemia/pathology , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , CDC28 Protein Kinase, S cerevisiae/physiology , Cell Survival/genetics , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Leukemic , Gene Rearrangement , Histone-Lysine N-Methyltransferase/metabolism , Humans , Mice , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Signal Transduction/genetics
2.
J Radiat Res ; 53(1): 72-8, 2012.
Article in English | MEDLINE | ID: mdl-22302047

ABSTRACT

PURPOSES: The Cks1 protein is a member of the highly conserved family of Cks/Suc1 proteins, which interact with Cdks, and was found to be an essential cofactor for efficient Skp2-dependent ubiquitination of p27. The present study was undertaken to examine the expression status of Cks1 in esophageal squamous cell carcinoma and its significance. MATERIALS AND METHODS: The expression of Cks1 in 140 esophageal squamous cell carcinoma patients was examined by immunohistochemistry. The correlations between Cks1 expression and tumor clinicopathologic features were analyzed. The effects of Cks1 expression on radiotherapy results were also examined. RESULTS: In the present study, we found that Cks1 is overexpressed in esophageal squamous cell carcinoma tissues. Elevated expression of Cks1 correlates significantly with tumor stage and positive lymph node metastasis (p < 0.05). Moreover, a significant negative correlation was found between Cks1 expression and the survival of patients who received radiotherapy (p < 0.05). At the molecular level, forced expression of Cks1 promotes the radio-resistance ability of EC9706 cells. Knockdown of Cks1 expression sensitizes cancer cells to radiation, and a wobble mutant of Cks1 that is resistant to Cks1 siRNA can rescue this effect. CONCLUSIONS: These results demonstrate for the first time that overexpression of Cks1 correlates with the increased radiotherapy resistance of esophageal squamous cell carcinoma.


Subject(s)
CDC2-CDC28 Kinases/physiology , Carcinoma, Squamous Cell/enzymology , Esophageal Neoplasms/enzymology , Neoplasm Proteins/physiology , Radiation Tolerance/physiology , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CDC2-CDC28 Kinases/antagonists & inhibitors , CDC2-CDC28 Kinases/biosynthesis , CDC2-CDC28 Kinases/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/radiotherapy , Carcinoma, Squamous Cell/surgery , Cell Line, Tumor/radiation effects , Cisplatin/administration & dosage , Combined Modality Therapy , Enzyme Induction , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/mortality , Esophageal Neoplasms/radiotherapy , Esophageal Neoplasms/surgery , Female , Fluorouracil/administration & dosage , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prognosis , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/physiology , Tumor Stem Cell Assay
3.
Exp Parasitol ; 112(3): 202-7, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16375894

ABSTRACT

The molecular mechanisms underlying gametocytogenesis in malaria parasites are not understood. Plasmodium falciparum cdc2-related kinase 1 (pfcrk-1), a gene that is expressed predominantly in gametocytes, bears homology to the PITSLRE subfamily of cyclin-dependent kinases and has been hypothesized to function as a negative regulator of the cell cycle. We attempted to knock-out pbcrk-1, the P. berghei orthologue of pfcrk-1, but were unable to recover P. berghei parasites with a disrupted pbcrk-1 locus. In contrast, an integration event at this locus that did not result in a loss-of-function of the pbcrk-1 gene was readily observed. This strongly suggests that a functional pbcrk-1 gene product is essential to intraerythrocytic asexual multiplication.


Subject(s)
CDC2-CDC28 Kinases/physiology , Erythrocytes/parasitology , Plasmodium berghei/enzymology , Plasmodium berghei/physiology , Amino Acid Sequence , Animals , Blotting, Northern , CDC2-CDC28 Kinases/chemistry , CDC2-CDC28 Kinases/genetics , Gene Deletion , Molecular Sequence Data , Plasmodium berghei/genetics , RNA, Protozoan/analysis , Rats , Reproduction, Asexual/physiology , Sequence Alignment
4.
Br J Haematol ; 130(6): 902-11, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16156860

ABSTRACT

The 3q21q26 syndrome leukaemias are characterised by dystrophic megakaryocytes, elevated platelet counts, ectopic EVI1 protein production and poor prognosis. To investigate the molecular basis of this disease, we developed a model system to examine the biological activity of EVI1 in a megakaryocyte progenitor cell line. For this purpose, Evi1 was conditionally expressed in human erythroleukaemia cells (HEL) that progress along the megakaryocyte lineage in the presence of 12-O-tetradecanoylphorbol 13-acetate (TPA). TPA-stimulated HEL cells normally undergo: (1) growth arrest; (2) altered morphology; (3) endomitosis and (4) characteristic changes in gene expression, including reduction of the erythroid-specific glycophoryn A and elevation of the specific glycoproteins GPIIIa and GPVI. Enforced Evi1 expression alone had no effect upon HEL cell proliferation or differentiation but a phenotype was manifest upon stimulation to differentiate. Evi1-expressing, TPA-treated HEL cells still showed growth arrest, had reduced and enhanced glycophoryn A and GPIIIa mRNA's, respectively, but failed to significantly elevate GPVI mRNA. This was accompanied by inhibition of endomitosis and altered cell morphology. Sustained CDK2 catalytic activity, typically associated with megakaryocyte endomitosis, was dramatically decreased in TPA-stimulated Evi1-expressing HEL cells because of significantly reduced levels of cyclin A. Therefore, enforced Evi1 expression could inhibit megakaryocyte differentiation although retention of some characteristic molecular changes, in combination with a block in endomitosis and altered morphology, suggest a defect in lineage progression. These results suggest that ectopic Evi1 expression contributes to a defective megakaryocyte differentiation programme and is likely to contribute to the phenotype observed in 3q21q26 syndrome leukaemias.


Subject(s)
CDC2-CDC28 Kinases/antagonists & inhibitors , DNA-Binding Proteins/physiology , Leukemia, Erythroblastic, Acute/pathology , Megakaryocytes/cytology , Proto-Oncogenes/physiology , Transcription Factors/physiology , CDC2-CDC28 Kinases/physiology , Cell Cycle , Cell Differentiation , Cell Proliferation , Cyclin-Dependent Kinase 2 , DNA-Binding Proteins/metabolism , Hematopoiesis , Humans , Leukemia, Erythroblastic, Acute/enzymology , Leukemia, Erythroblastic, Acute/metabolism , MDS1 and EVI1 Complex Locus Protein , Megakaryocytes/enzymology , Mitosis , Neoplasm Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/metabolism , Tumor Cells, Cultured
5.
Cancer Cell ; 7(6): 591-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15950907

ABSTRACT

p27(Kip1) and p21(Cip1) are thought to suppress tumor growth and prevent cell cycle progression by inhibiting Cdk2-cyclin E/A kinases. Since Cdk2 is dispensable for mitotic cell division, we analyzed the activity of these inhibitors in Cdk2-deficient cells. Ectopic expression of p27(Kip1) or p21(Cip1) efficiently inhibits cell cycle progression of Cdk2(-/-) fibroblasts. Loss of p27(Kip1) or p21(Cip1) confers similar proliferative advantages to Cdk2(+/+) and Cdk2(-/-) cells. Moreover, Cdk2 is dispensable for p21(Cip1)-induced cell cycle arrest after DNA damage. Finally, ablation of Cdk2 in p27(Kip1) null mice does not suppress their phenotypic defects, including development of pituitary tumors. These results indicate that Cdk2 is not an essential target for p27(Kip1) and p21(Cip1) in cell cycle inhibition and tumor suppression.


Subject(s)
Cell Cycle Proteins/physiology , Cell Cycle/physiology , Tumor Suppressor Proteins/physiology , Animals , Body Weight/genetics , CDC2-CDC28 Kinases/genetics , CDC2-CDC28 Kinases/metabolism , CDC2-CDC28 Kinases/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , DNA Damage , Embryo, Mammalian/cytology , Etoposide/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Hyperplasia , Mice , Mice, Knockout , Mice, Nude , Mutation , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Retinal Dysplasia/genetics , Retinal Dysplasia/pathology , Retroviridae/genetics , Transfection , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
J Biol Chem ; 280(23): 21731-8, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15824106

ABSTRACT

Cyclin-dependent kinases (CDKs) are a family of evolutionarily conserved serine/threonine kinases. CDK2 acts as a checkpoint for the G(1)/S transition in the cell cycle. Despite a down-regulation of CDK2 activity in postmitotic cells, many cell types, including muscle cells, maintain abundant levels of CDK2 protein. This led us to hypothesize that CDK2 may have a function in postmitotic cells. We show here for the first time that CDK2 can be activated by neuregulin (NRG) in differentiated C2C12 myotubes. In addition, this activity is required for expression of the acetylcholine receptor (AChR) epsilon subunit. The switch from the fetal AChRgamma subunit to the adult-type AChRepsilon is required for synapse maturation and the neuromuscular junction. Inhibition of CDK2 activity with either the specific CDK2 inhibitory peptide Tat-LFG or by RNA interference abolished neuregulin-induced AChRepsilon expression. Neuregulin-induced activation of CDK2 also depended on the ErbB receptor, MAPK, and PI3K, all of which have previously been shown to be required for AChRepsilon expression. Neuregulin regulated CDK2 activity through coordinating phosphorylation of CDK2 on Thr-160, accumulation of CDK2 in the nucleus, and down-regulation of the CDK2 inhibitory protein p27 in the nucleus. In addition, we also observed a novel mechanism of regulation of CDK2 activity by a low molecular weight variant of cyclin E in response to NRG. These findings establish CDK2 as an intermediate molecule that integrates NRG-activated signals from both the MAPK and PI3K pathways to AChRepsilon expression and reveal an undiscovered physiological role for CDK2 in postmitotic cells.


Subject(s)
CDC2-CDC28 Kinases/physiology , Muscle Fibers, Skeletal/metabolism , Neuregulin-1/metabolism , Receptors, Cholinergic/metabolism , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/physiology , Animals , Blotting, Western , CDC2-CDC28 Kinases/metabolism , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , DNA Primers/chemistry , Down-Regulation , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Immunoprecipitation , MAP Kinase Signaling System , Mice , Mitosis , Muscles/metabolism , Oncogene Proteins v-erbB/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , RNA/metabolism , RNA Interference , Recombinant Proteins/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Subcellular Fractions , Synapses/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
7.
J Biol Chem ; 280(18): 18095-107, 2005 May 06.
Article in English | MEDLINE | ID: mdl-15741163

ABSTRACT

By having demonstrated previously that p27(Kip1), a potent inhibitor of G(1) cyclin-cyclin-dependent kinases complexes, increases markedly during intestinal epithelial cell differentiation, we examined the effect of p27(Kip1) on the activity of the transcription factor CDX2. The present results revealed the following. 1) p27(Kip1) interacts with the CDX2 transcription factor. 2) In contrast to CDX2 mRNA levels, CDX2 protein expression levels significantly increased as soon as Caco-2/15 cells reached confluence, slowed their proliferation, and began their differentiation. The mechanism of CDX2 regulation is primarily related to protein stability, because inhibition of proteasome activity increased CDX2 levels. The half-life of CDX2 protein was significantly enhanced in differentiated versus undifferentiated proliferative intestinal epithelial cells. 3) Cdk2 interacted with CDX2 and phosphorylated CDX2, as determined by pull-down glutathione S-transferase and immunoprecipitation experiments with proliferating undifferentiated Caco-2/15 cell extracts. 4) Treatment of Caco-2/15 cells with MG132 (a proteasome inhibitor) and (R)-roscovitine (a specific Cdk2 inhibitor) induced an increase in CDX2 protein levels. 5) Conversely, ectopic expression of Cdk2 resulted in decreased expression of CDX2 protein. 6) Of note, treatment of proliferative Caco-2/15 cells with (R)-roscovitine or leptomycin (an inhibitor of nuclear export through CRM1) led to an accumulation of CDX2 into the nucleus. These data suggest that CDX2 undergoes CRM1-dependent nuclear export and cytoplasmic degradation in cells in which Cdk2 is activated, such as in proliferative intestinal epithelial cells. The targeted degradation of CDX2 following its phosphorylation by Cdk2 identifies a new mechanism through which CDX2 activity can be regulated in coordination with the cell cycle machinery.


Subject(s)
Active Transport, Cell Nucleus/physiology , CDC2-CDC28 Kinases/physiology , Homeodomain Proteins/metabolism , Intestinal Mucosa/metabolism , Proteasome Endopeptidase Complex/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , CDC2-CDC28 Kinases/genetics , CDX2 Transcription Factor , Caco-2 Cells , Cricetinae , Cyclin-Dependent Kinase 2 , Homeodomain Proteins/genetics , Humans , Intestinal Mucosa/drug effects , Leupeptins/pharmacology , Mice , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors
8.
J Reprod Dev ; 51(1): 143-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15750306

ABSTRACT

The meiotic progression of Xenopus oocytes has been suggested to depend on the activity of cyclin-dependent kinase 2 (Cdk2). We examined whether Cdk2 is involved in the regulation of mammalian oocyte meiosis by injecting porcine oocytes with anti-Cdk2 antibody. At first, the cross-reactivity of the anti-Cdk2 antibody with Cdc2 kinase was evaluated by immunoprecipitation and immunoblotting experiments using porcine granulosa cell extract, and no cross-reactivity with Cdc2 kinase was observed in the antibody used. In the anti-Cdk2 antibody-injected group, 50.7% of the oocytes were arrested in the second metaphase after 50 h of culture and this rate was significantly lower than those in the non-injected intact oocytes or the oocytes injected with mouse IgG (84.5% and 86.7%, respectively). Most of the other oocytes in the antibody-injected group formed a pronucleus without polar bodies or with only one polar body. The cyclin B1 amount in the antibody-injected and activated oocytes was dramatically decreased compared with that in the intact or mouse IgG-injected oocytes after 50 h of culture. These results suggest that Cdk2 is involved in the meiotic maturation of mammalian oocytes, and that the block of Cdk2 activity results in the failure of cyclin B1 accumulation and second meiosis induction.


Subject(s)
CDC2-CDC28 Kinases/physiology , Oocytes/metabolism , Animals , CDC2-CDC28 Kinases/metabolism , Cyclin B/metabolism , Cyclin B1 , Cyclin-Dependent Kinase 2 , Female , Granulosa Cells/cytology , Granulosa Cells/metabolism , Immunoblotting , Immunoglobulin G/chemistry , Immunoprecipitation , Meiosis , Oogenesis , Swine , Time Factors , Xenopus , Xenopus Proteins
9.
Genes Dev ; 19(1): 114-26, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15598982

ABSTRACT

In eukaryotes, prereplication complexes (pre-RCs) containing ORC, Cdc6, Cdt1, and MCM2-7 are assembled on chromatin in the G1 phase. In S phase, when DNA replication initiates, pre-RCs are disassembled, and new pre-RC assembly is restricted until the following G1 period. As a result, DNA replication is limited to a single round per cell cycle. One inhibitor of pre-RC assembly, geminin, was discovered in Xenopus, and it binds and inactivates Cdt1 in S phase. However, removal of geminin from Xenopus egg extracts is insufficient to cause rereplication, suggesting that other safeguards against rereplication exist. Here, we show that Cdt1 is completely degraded by ubiquitin-mediated proteolysis during the course of the first round of DNA replication in Xenopus egg extracts. Degradation depends on Cdk2/Cyclin E, Cdc45, RPA, and polymerase alpha, demonstrating a requirement for replication initiation. Cdt1 is ubiquitinated on chromatin, and this process also requires replication initiation. Once replication has initiated, Cdk2/Cyclin E is dispensable for Cdt1 degradation. When fresh Cdt1 is supplied after the first round of DNA replication, significant rereplication results, and rereplication is enhanced in the absence of geminin. Our results identify a replication-dependent proteolytic pathway that targets Cdt1 and that acts redundantly with geminin to inactivate Cdt1 in S phase.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/genetics , DNA Replication , DNA-Binding Proteins/metabolism , Animals , CDC2-CDC28 Kinases/physiology , Cell Cycle Proteins/physiology , Cell Extracts , Chromatin , Cyclin-Dependent Kinase 2 , DNA Polymerase I , Ovum/cytology , Peptide Hydrolases/metabolism , S Phase , Ubiquitin , Xenopus , Xenopus Proteins
10.
Cancer Cell ; 6(6): 565-76, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15607961

ABSTRACT

The genomic organization of the CDK2 gene, which overlaps the melanocyte-specific gene SILV/PMEL17, poses an interesting regulatory challenge. We show that, despite its ubiquitous expression, CDK2 exhibits tissue-specific regulation by the essential melanocyte lineage transcription factor MITF. In addition, functional studies revealed this regulation to be critical for maintaining CDK2 kinase activity and growth of melanoma cells. Expression levels of MITF and CDK2 are tightly correlated in primary melanoma specimens and predict susceptibility to the CDK2 inhibitor roscovitine. CDK2 depletion suppressed growth and cell cycle progression in melanoma, but not other cancers, corroborating previous results. Collectively, these data indicate that CDK2 activity in melanoma is largely maintained at the transcriptional level by MITF, and unlike other malignancies, it may be a suitable drug target in melanoma.


Subject(s)
CDC2-CDC28 Kinases/physiology , DNA-Binding Proteins/physiology , Melanoma/pathology , Transcription Factors/physiology , Blotting, Western , CDC2-CDC28 Kinases/genetics , CDC2-CDC28 Kinases/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E-Box Elements/physiology , Fibroblasts/metabolism , Flow Cytometry , Gene Expression , Gene Expression Regulation, Neoplastic , Genes, Reporter/genetics , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/metabolism , Membrane Glycoproteins , Microphthalmia-Associated Transcription Factor , Mutation , Oligonucleotide Array Sequence Analysis , Protein Kinase Inhibitors/pharmacology , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Purines/pharmacology , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Roscovitine , S Phase/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transfection , bcl-X Protein , gp100 Melanoma Antigen
11.
J Biol Chem ; 279(53): 55348-54, 2004 Dec 31.
Article in English | MEDLINE | ID: mdl-15513912

ABSTRACT

Fibroblast growth factors (FGFs) are upstream activators of the mitogen-activated protein kinase pathway and mitogens in a wide variety of cells. However, whether the mitogen-activated protein kinase pathway solely accounts for the induction of cell cycle or antiapoptotic activity of the FGF receptor (FGFR) tyrosine kinase is not clear. Here we report that cell cycle inducer Cks1, which triggers ubiquitination and degradation of p27(Kip1), associates with the unphosphorylated form of FGFR substrate 2 (FRS2), an adaptor protein that is phosphorylated by FGFR kinases and recruits downstream signaling molecules. FGF-dependent activation of FGFR tyrosine kinases induces FRS2 phosphorylation, causes release of Cks1 from FRS2, and promotes degradation of p27(Kip1) in 3T3 cells. Since degradation of p27(Kip1) is a key regulatory step in activation of the cyclin E/A-Cdk complex during the G(1)/S transition of the cell cycle, the results suggest a novel mitogenic pathway whereby FGF and other growth factors that activate FRS2 directly activate cyclin-dependent kinases.


Subject(s)
CDC2-CDC28 Kinases/physiology , Membrane Proteins/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Fibroblast Growth Factor/physiology , 3T3 Cells , Animals , CDC2-CDC28 Kinases/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p27 , DNA, Complementary/metabolism , G1 Phase , Glutathione/metabolism , Glutathione Transferase/metabolism , Growth Substances/metabolism , MAP Kinase Signaling System , Mice , Models, Biological , Mutation , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/chemistry , S Phase , Sepharose/chemistry , Signal Transduction , Time Factors , Tumor Suppressor Proteins/metabolism , Tyrosine/metabolism , Ubiquitin/metabolism
12.
DNA Cell Biol ; 23(8): 475-89, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15307950

ABSTRACT

The centrosome is a unique organelle that functions as the microtubule organizing center in most animal cells. During cell division, the centrosomes form the poles of the bipolar mitotic spindle. In addition, the centrosomes are also needed for cytokinesis. Each mammalian somatic cell typically contains one centrosome, which is duplicated in coordination with DNA replication. Just like the chromosomes, the centrosome is precisely reproduced once and only once during each cell cycle. However, it remains a mystery how this protein-based structure undergoes accurate duplication in a semiconservative manner. Intriguingly, amplification of the centrosome has been found in numerous forms of cancers. Cells with multiple centrosomes tend to form multipolar spindles, which result in abnormal chromosome segregation during mitosis. It has therefore been postulated that centrosome aberration may compromise the fidelity of cell division and cause chromosome instability. Here we review the current understanding of how the centrosome is assembled and duplicated. We also discuss the possible mechanisms by which centrosome abnormality contributes to the development of malignant phenotype.


Subject(s)
Centrosome/physiology , Mitosis/physiology , Models, Biological , Spindle Apparatus/physiology , Aneuploidy , Aurora Kinases , CDC2-CDC28 Kinases/physiology , Cell Cycle Proteins , Cell Division/physiology , Cell Transformation, Neoplastic/pathology , Chromosomal Instability/physiology , Cyclin-Dependent Kinase 2 , DNA Replication/physiology , Protein Serine-Threonine Kinases , Protein Tyrosine Phosphatases , Saccharomyces cerevisiae Proteins
13.
Mol Cell Biol ; 24(13): 6058-66, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15199159

ABSTRACT

Ubiquitin-dependent proteolysis makes a major contribution to decreasing the levels of p27. Ubiquitin-dependent proteolysis of p27(kip1) is growth and cell cycle regulated in two ways: first, skp2, a component of the E3-ubiquitin ligase, is growth regulated, and second, a kinase must phosphorylate the threonine-187 position on p27 so that it can be recognized by skp2. In vitro, p27 is phosphorylated by cyclin E- and cyclin A-associated cdk2 as well as by cyclin B1-cdk1. Having analyzed the effect of different cyclin-cyclin-dependent kinase complexes on ubiquitination of p27 in a reconstitution assay system, we now report a noncatalytic requirement for cyclin A-cdk2. Multiparameter flow cytometric analysis also indicates that p27 turnover correlates best with the onset of S phase, once the levels of cyclin A become nearly maximal. Finally, increasing the amount of both cyclin E-cdk2 and skp2 was less efficient at promoting p27 ubiquitination than was increasing the amount of cyclin A-cdk2 alone in extracts prepared from cultures of >93%-purified G(1) cells. Together these lines of evidence suggest that cyclin A-cdk2 plays an ancillary noncatalytic role in the ubiquitination of p27 by the SCF(skp2) complex.


Subject(s)
CDC2-CDC28 Kinases/physiology , Cell Cycle Proteins/metabolism , Cyclin A/physiology , Tumor Suppressor Proteins/metabolism , CDC2-CDC28 Kinases/analysis , Cyclin A/analysis , Cyclin A/genetics , Cyclin E/analysis , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Flow Cytometry , G1 Phase , HeLa Cells , Humans , Mutation , S Phase , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin/metabolism
14.
Int J Biochem Cell Biol ; 36(8): 1424-39, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15147722

ABSTRACT

E-type cyclins (cyclin E1 and cyclin E2) are expressed during the late G1 phase of the cell cycle until the end of the S-phase. The activity of cyclin E is limiting for the passage of cells through the restriction point "R" which marks a "point of no return" for cells entering the division cycle from a resting state or passing from G1 into S-phase. Expression of cyclin E is regulated on the level of gene transcription mainly by members of the E2F trrnscription factor family and by its degradation via the proteasome pathway. Cyclin E binds and activates the kinase Cdk2 and by phosphorylating its substrates, the so-called "pocket proteins", the cyclic/Cdk2 complexes initiate a cascade of events that leads to the expression of S-phase specific genes. Aside from this specific function as a regulator of S-phase-entry, cyclin E plays a direct role in the initiation of DNA replication, the control of genomic stability, and the centrosome cycle. Surprisingly, recent studies have shown that the once thought essential cyclin E is dispensable for the development of higher eukaryotes and for the mitotic division of eukaryotic cells. Nevertheless, high level cyclin E expression has been associated with the initiation or progression of different human cancers, in particular breast cancer but also leukemia, lymphoma and others. Transgenic mouse models in which cyclin E is constitutively expressed develop malignant diseases, supporting the notion of cyclin E as a dominant onco-protein.


Subject(s)
Cyclin E , Animals , CDC2-CDC28 Kinases/metabolism , CDC2-CDC28 Kinases/physiology , Cyclin E/chemistry , Cyclin E/genetics , Cyclin E/metabolism , Cyclin E/physiology , Cyclin-Dependent Kinase 2 , Humans , Interphase , Neoplasms/etiology , Ubiquitin/metabolism
15.
Sci STKE ; 2004(224): pe11, 2004 Mar 09.
Article in English | MEDLINE | ID: mdl-15026579

ABSTRACT

The canonical view of the mammalian cell cycle arose from studies of cultured cells rather than mutant organisms. It depicts the many complexes of cyclin and Cdk (cyclin/Cdk) as fulfilling unique and essential steps that dictate the sequential order of cell cycle events. Recent analyses of knockout mice challenge this view. Cdk2 and cyclin E, long thought to be essential, are largely dispensable. Here, we discuss the phenotypes of these and other cyclin/Cdk mutants in genetically tractable metazoa (mouse, fly, and nematode) and explore possible reasons behind similarities and differences among experimental systems and cell types.


Subject(s)
CDC2-CDC28 Kinases/physiology , Cell Cycle/physiology , Cyclin E/physiology , Animals , CDC2-CDC28 Kinases/deficiency , CDC2-CDC28 Kinases/genetics , Cells, Cultured , Cyclin E/deficiency , Cyclin E/genetics , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA Replication , Embryonic and Fetal Development/physiology , Gene Targeting , Humans , Invertebrates/genetics , Invertebrates/metabolism , Macromolecular Substances , Mammals/genetics , Mammals/metabolism , Mice , Mice, Knockout , Models, Biological , Phenotype , Phosphorylation , Protein Processing, Post-Translational , Retinoblastoma Protein/physiology , S Phase/physiology , Substrate Specificity
16.
Circ Res ; 94(8): 1067-74, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-15031262

ABSTRACT

Proteoglycans are produced and secreted by vascular smooth muscle cells, but the pathophysiological role of these glycoproteins in the vasculature is an enigma. Because the small leucine-rich proteoglycan (SLRP) biglycan is overexpressed in arteriosclerotic lesions, we produced mice constitutively overexpressing biglycan in the vascular smooth muscle, in order to examine the effects on vascular pathology. In the aorta and renal vasculature, increased vascular proliferation was seen both in the basal state and after infusion of angiotensin II (Ang II) in the transgenic mice compared with wild-type controls. In addition, the combination of biglycan overexpression and Ang II infusion resulted in marked increases in vascular smooth muscle cell proliferation and migration in the coronary arteries, as well as increases in fibrosis surrounding the vessels. In vitro, biglycan caused an increase in thymidine incorporation and migration of vascular smooth muscle cells, whereas these parameters were unchanged or reduced in endothelial cells. Moreover, addition of biglycan resulted in an increase in cdk2 expression and decrease in p27 levels in the vascular smooth muscle cells. These results suggest that this extracellular matrix SLRP may be involved in the regulation of vascular smooth muscle growth and migration through cdk2- and p27-dependent pathways. Furthermore, changes in biglycan expression could be a factor influencing the susceptibility of arteries to vascular injury, and may play a direct role in the pathogenesis of vascular lesions.


Subject(s)
Arterial Occlusive Diseases/etiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Proteoglycans/physiology , Actins/genetics , Angiotensin II/genetics , Angiotensin II/pharmacology , Animals , Aorta/metabolism , Aorta/ultrastructure , Arterial Occlusive Diseases/genetics , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/pathology , Arterioles/metabolism , Arterioles/ultrastructure , Biglycan , CDC2-CDC28 Kinases/genetics , CDC2-CDC28 Kinases/physiology , Cattle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Division , Cell Movement , Cells, Cultured/cytology , Cells, Cultured/metabolism , Coronary Vessels/metabolism , Coronary Vessels/ultrastructure , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Disease Susceptibility , Endothelial Cells/cytology , Extracellular Matrix Proteins , Gene Expression Regulation , Humans , Kidney/blood supply , Male , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/injuries , Myocytes, Smooth Muscle/physiology , Organ Specificity , Promoter Regions, Genetic/genetics , Proteoglycans/biosynthesis , Proteoglycans/genetics , Rats , Rats, Wistar , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/physiology , Renin/blood , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
17.
Oncogene ; 23(19): 3361-7, 2004 Apr 22.
Article in English | MEDLINE | ID: mdl-14767478

ABSTRACT

Mitotic entry, a critical decision point for maintaining genetic stability, is governed by the cyclin B/Cyclin dependent kinase 1 (Cdc2) complex. In Xenopus oocytes and early embryos, accumulation of cyclin B activates Cdk1, which then phosphorylates and activates the positive regulator Cdc25 in an autocatalytic feedback loop. However, cyclin B levels do not increase as some human cells approach mitosis, and the key factors regulating Cdk1 activation in human cells are unknown. We report here that reducing cyclin A expression by RNA interference (RNAi) in primary human fibroblasts inhibited activation of Cdc25B and Cdc25C and dephosphorylation of Cdk1 on tyrosine (tyr) 15. These results were reproduced in U2-OS cells by inducing the expression of a dominant-negative (dn) mutant of Cdk2, the principal cyclin A binding partner. Cdk2-dn induction could inhibit Cdc25B activity and foster Cdk1 tyr phosphorylation within the S phase, temporally dissociating these events from Cdk1 activation at mitosis. In contrast, reducing Cdk1 expression delayed mitotic entry without markedly impairing Cdc25B or Cdc25C activity. These results suggest that cyclin A/Cdk2 complexes are key regulators of Cdc25 and Cdk1 activation in human cells. This pathway appears to be commonly deregulated in cancer.


Subject(s)
CDC2 Protein Kinase/metabolism , CDC2-CDC28 Kinases/physiology , Cyclin A/physiology , cdc25 Phosphatases/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 2 , Enzyme Activation , G2 Phase , Humans , Phosphorylation , S Phase
18.
Biol Reprod ; 70(4): 1147-52, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14695906

ABSTRACT

In all systems examined so far, the G2/M phase transition is controlled by the M-phase promoting factor (MPF), a complex of cdc2 (CDK1) and cyclin B1. Histone H1 kinase activity and MPF components are present in pachytene spermatocytes (PS). However, it has not been demonstrated yet that direct inhibition of MPF activity prevents the G2/M transition in these cells. When roscovitine, a potent inhibitor of CDK1, CDK2, and CDK5 activities, was added to cocultures of PS with Sertoli cells, the number of both secondary spermatocytes and round spermatids formed were lower than in control cultures, despite similar cell viability. This effect of roscovitine was reversible, did not involve the Sertoli cells, and was dependent on the concentration of the inhibitor. Roscovitine did not modify the amount of MPF in these germ cells but inhibited the CDK1- or CDK2-associated histone H1 kinase activity of PS. Hence a functional relationship between cyclin-dependent kinase activity and the spontaneous processing of the first meiotic division and, for the first time, of the second meiotic division of male germ cells is shown.


Subject(s)
CDC2 Protein Kinase/physiology , CDC2-CDC28 Kinases/physiology , Cyclin-Dependent Kinases/physiology , Spermatocytes/cytology , Spermatocytes/enzymology , Animals , CDC2 Protein Kinase/metabolism , CDC2-CDC28 Kinases/metabolism , Cell Size , Cell Survival , Cells, Cultured , Coculture Techniques , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 5 , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Male , Maturation-Promoting Factor/metabolism , Meiosis/physiology , Osmolar Concentration , Protein Kinase Inhibitors , Protein Kinases/metabolism , Purines/administration & dosage , Purines/pharmacology , Rats , Roscovitine , Sertoli Cells , Spermatids/cytology , Spermatids/physiology , Spermatocytes/physiology
19.
Cancer Lett ; 202(2): 201-11, 2003 Dec 30.
Article in English | MEDLINE | ID: mdl-14643450

ABSTRACT

Human PRL-1, PRL-2, and PRL-3 tyrosine phosphatases induce the malignant transformation of epithelial cells. We tested the hypothesis that the oncogenic effects of PRL occur by increasing cellular proliferation. Cells stably transfected with PRL-1 or PRL-2 exhibited 2.7-3.3-fold increases over control cells in the rate of DNA synthesis and the proportion of cells in S-phase, and they progressed more rapidly from G1 into S. In addition, cells overexpressing either PRL-1 or PRL-2 exhibited enhanced cyclin-dependent kinase 2 (CDK2) activity and significantly lower p21(Cip1/Waf1) protein levels, and PRL-1 overexpressing cells had higher cyclin A protein levels than control cells. We conclude that PRL phosphatases increase cell proliferation by stimulating progression from G1 into S phase, and this process may be dependent on the down regulation of the cyclin dependent kinase inhibitor p21(Cip1/Waf1).


Subject(s)
Cell Cycle/physiology , Cyclins/physiology , Protein Tyrosine Phosphatases/physiology , Animals , Apoptosis/physiology , CDC2-CDC28 Kinases/physiology , Cell Division/physiology , Cells, Cultured , Cricetinae , Cyclin A/physiology , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Down-Regulation , Humans , Immunoblotting , Polymerase Chain Reaction , Precipitin Tests , Transfection
20.
Mol Cell ; 12(2): 381-92, 2003 Aug.
Article in English | MEDLINE | ID: mdl-14536078

ABSTRACT

Autophosphorylation-triggered ubiquitination has been proposed to be the major pathway regulating cyclin E protein abundance: phosphorylation of cyclin E on T380 by its associated CDK allows binding to the receptor subunit, Fbw7, of the SCFFbw7 ubiquitin ligase. We have tested this model in vivo and found it to be an inadequate representation of the pathways that regulate cyclin E degradation. We show that assembly of cyclin E into cyclin E-Cdk2 complexes is required in vivo for turnover by the Fbw7 pathway; that Cdk2 activity is required for cyclin E turnover in vivo because it phosphorylates S384; that phosphorylation of T380 in vivo does not require Cdk2 and is mediated primarily by GSK3; and that two additional phosphorylation sites, T62 and S372, are also required for turnover. Thus, cyclin E turnover is controlled by multiple biological inputs and cannot be understood in terms of autophosphorylation alone.


Subject(s)
CDC2-CDC28 Kinases/physiology , Cyclin E/metabolism , Glycogen Synthase Kinase 3/physiology , Animals , Binding Sites , CDC2-CDC28 Kinases/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase 2 , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinases/metabolism , Humans , Immunoblotting , Mice , Mutation , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Retroviridae/metabolism , Time Factors , Transfection , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...