Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Curr Top Dev Biol ; 120: 1-53, 2016.
Article in English | MEDLINE | ID: mdl-27475848

ABSTRACT

The preimplantation development stage of mammalian embryogenesis consists of a series of highly conserved, regulated, and predictable cell divisions. This process is essential to allow the rapid expansion and differentiation of a single-cell zygote into a multicellular blastocyst containing cells of multiple developmental lineages. This period of development, also known as the germinal stage, encompasses several important developmental transitions, which are accompanied by dramatic changes in cell cycle profiles and dynamics. These changes are driven primarily by differences in the establishment and enforcement of cell cycle checkpoints, which must be bypassed to facilitate the completion of essential cell cycle events. Much of the current knowledge in this area has been amassed through the study of knockout models in mice. These mouse models are powerful experimental tools, which have allowed us to dissect the relative dependence of the early embryonic cell cycles on various aspects of the cell cycle machinery and highlight the extent of functional redundancy between members of the same gene family. This chapter will explore the ways in which the cell cycle machinery, their accessory proteins, and their stimuli operate during mammalian preimplantation using mouse models as a reference and how this allows for the usually well-defined stages of the cell cycle to be shaped and transformed during this unique and critical stage of development.


Subject(s)
Cell Cycle , Embryo, Mammalian/cytology , Embryonic Development , Animals , Cell Cycle Checkpoints , DNA Damage , Humans , Models, Animal
2.
Oncogene ; 34(7): 932-8, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-24531709

ABSTRACT

Recent studies suggest that physiological and tumorigenic proliferation of mammalian cells is controlled by multiple cyclin-dependent kinases (CDKs) largely in tissue-specific manners. We and others previously demonstrated that adult mice deficient for the Cyclin D partner CDK4 (Cdk4(-/-) mice) exhibit hypoplasia in the pituitary and pancreatic islet due to primary postnatal defects in proliferation. Intriguingly, those neuroendocrine tissues affected in Cdk4(-/-) mice are the primary targets of tumorigenesis in the syndrome of multiple endocrine neoplasia type-1 (MEN1). Mice with heterozygous disruption of the tumor suppressor Men1 gene (Men1(+/-)) develop tumors in the pituitary, pancreatic islets and other neuroendocrine tissues, which is analogous to humans with MEN1 mutations. To explore the genetic interactions between loss of Men1 and activation of CDKs, we examined the impact of Cdk4 or Cdk2 disruption on tumorigenesis in Men1(+/-) mice. A majority of Men1(+/-) mice with wild-type CDKs developed pituitary and islet tumors by 15 months of age. Strikingly, Men1(+/-); Cdk4(-/-) mice did not develop any tumors, and their islets and pituitaries remained hypoplastic with decreased proliferation. In contrast, Men1(+/-); Cdk2(-/-) mice showed pituitary and islet tumorigenesis comparable to those in Men1(+/-) mice. Pituitaries of Men1(+/-); Cdk4(-/-) mice showed no signs of loss of heterozygosity (LOH) in the Men1 locus, whereas tumors in Men1(+/-) mice and Men1(+/-); Cdk2(-/-) mice exhibited LOH. Consistently, CDK4 knockdown in INS-1 insulinoma cells inhibited glucose-stimulated cell cycle progression with a significant decrease in phosphorylation of retinoblastoma protein (RB) at specific sites including Ser780. CDK2 knockdown had minimum effects on RB phosphorylation and cell cycle progression. These data suggest that CDK4 is a critical downstream target of MEN1-dependent tumor suppression and is required for tumorigenic proliferation in the pituitary and pancreatic islet, whereas CDK2 is dispensable for tumorigenesis in these neuroendocrine cell types.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Insulinoma/metabolism , Pancreatic Neoplasms/metabolism , Pituitary Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 4/genetics , Female , Humans , Insulinoma/genetics , Insulinoma/pathology , Male , Mice , Mice, Knockout , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation/genetics , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Rats , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Transcription Factors/genetics
3.
Cell Death Differ ; 17(9): 1486-500, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20203688

ABSTRACT

Chemotherapeutics (e.g., aurora kinase inhibitors) designed to target proliferative cells are often nonspecific for tumor cells as normal cycling cells are also susceptible. Indeed, one of the major dose-limiting toxicities of aurora kinase inhibitors is a dangerous depletion of neutrophils in patients. In this study we proposed a strategy to selectively target p53 mutant cells while sparing normal ones. The strategy is based on the understanding that normal cells have an intact p53 pathway but not tumor cells carrying p53 mutations. Nongenotoxic activation of p53 using nutlin led to a reversible activation of G1 and G2 arrest in normal cells, which prevents them from entering mitosis, thus protecting them from the side effects of aurora kinase inhibition (VX-680), namely endoreduplication and apoptosis. Cells carrying mutant p53 are selectively killed by the nutlin/VX-680 combination, whereas p53 wild-type cells retain their proliferative capacity. The major implications drawn from these results are: (1) reversible nongenotoxic activation of p53 may be used as a strategy for the chemoprotection of normal tissues, and (2) aurora kinase inhibitors may have alleviated side effects when used in combination with nutlin-like inhibitors. We highlight the distinct roles of p53 and p73 in mediating the cellular responses to VX-680 and suggest that dual protection by p53 and p73 are needed to guard against endoreduplication and polyploidy.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Mutation/physiology , Piperazines/pharmacology , Tumor Suppressor Protein p53/genetics , Apoptosis/genetics , Aurora Kinases , Caffeine/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Cyclin A2/genetics , Cyclin A2/metabolism , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/genetics , G2 Phase/drug effects , Gene Expression/drug effects , Gene Expression/genetics , Humans , Imidazoles/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/genetics , Piperazines/therapeutic use , Polyploidy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , RNA, Small Interfering/genetics , Tetraploidy , Tumor Protein p73 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
4.
Oncogene ; 28(33): 2925-39, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19561645

ABSTRACT

After a decade of extensive work on gene knockout mouse models of cell-cycle regulators, the classical model of cell-cycle regulation was seriously challenged. Several unexpected compensatory mechanisms were uncovered among cyclins and Cdks in these studies. The most astonishing observation is that Cdk2 is dispensable for the regulation of the mitotic cell cycle with both Cdk4 and Cdk1 covering for Cdk2's functions. Similar to yeast, it was recently discovered that Cdk1 alone can drive the mammalian cell cycle, indicating that the regulation of the mammalian cell cycle is highly conserved. Nevertheless, cell-cycle-independent functions of Cdks and cyclins such as in DNA damage repair are still under investigation. Here we review the compensatory mechanisms among major cyclins and Cdks in mammalian cell-cycle regulation.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Animals , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/metabolism , DNA Damage , Humans , Meiosis , Mice , Mice, Knockout , Models, Biological
5.
Oncogene ; 26(31): 4469-77, 2007 Jul 05.
Article in English | MEDLINE | ID: mdl-17297466

ABSTRACT

Inactivation of cyclin-dependent kinases (Cdks) and/or cyclins in mice has changed our view of cell cycle regulation. In general, cells are far more resistant to the loss of Cdks than originally anticipated, suggesting widespread compensation among the Cdks. Early embryonic cells are, so far, not sensitive to the lack of multiple Cdks or cyclins. In contrast, differentiated cells are more dependent on Cdk/cyclin complexes and the functional redundancy is more limited. Our challenge is to better understand these cell-type specific differences in cell cycle regulation that can be used to design efficient cancer therapy. Indeed, tumor cells seem to respond to inhibition of Cdk activities, however, with different outcome depending on the tumor cell type. Tumor cells share some proliferation features with stem cells, but appear more sensitive to loss of Cdk activity, somewhat resembling differentiated cells. We summarize the current knowledge of cell cycle regulation in different cell types and highlight their sensitivity to the lack of Cdk activities.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinases/deficiency , Neoplasms/enzymology , Animals , Apoptosis , Cell Death , Cell Differentiation , Cell Line, Tumor , Cyclins , Embryonic Stem Cells/physiology , Gene Silencing , Mice , Mutation , Signal Transduction
6.
Clin Exp Immunol ; 145(1): 139-46, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16792684

ABSTRACT

The objective of this study was to investigate the occurrence of apoptosis of monocytes in an experimental model of multiple trauma and its probable correlation to bacterial translocation. Thirty-two rabbits were applied in three groups: A, controls; B, myotomy of the right femur; and C, myotomy and fracture of the right femur. Blood was sampled for the estimation of endotoxins [lipopolysaccharide (LPS)], tumour necrosis factor (TNF)-alpha, malondialdehyde (MDA) and isolation of peripheral blood mononuclear cells (PBMCs). PBMCs, derived after centrifugation over Ficoll, were incubated in flasks and apoptosis of non-adherent lymphocytes and adherent monocytes was estimated after staining for Annexin-V and flow cytometry. TNF-alpha of supernatants of cultured monocytes was also determined. Tissue segments were cultured after death. Median survival of groups A, B and C was > 14, > 14 and 9.00 days, respectively. Apoptosis of lymphocytes in group C was higher than group A at 2, 4 and 48 h and of monocytes in group C higher than group A at 2 and 4 hours. LPS in group C was higher than group A at 2, 4 and 48 h. Apoptosis of lymphocytes and monocytes was correlated positively with serum TNF-alpha and negatively with TNF-alpha of monocyte supernatants. Cultures of organ segments of group A were sterile. Pseudomonas aeruginosa was isolated from liver, lung and spleen in five animals in group B (45.45%) and in six in group C (54.54%). Early apoptosis of blood monocytes supervened after multiple trauma; the phenomenon was accompanied by apoptosis of blood lymphocytes and subsequent bacterial translocation.


Subject(s)
Lymphocytes/pathology , Monocytes/pathology , Multiple Trauma/immunology , Systemic Inflammatory Response Syndrome/immunology , Animals , Apoptosis , Bacterial Translocation , Cells, Cultured , Fractures, Bone/immunology , Lipopolysaccharides/blood , Male , Malondialdehyde/blood , Models, Animal , Multiple Trauma/microbiology , Rabbits , Systemic Inflammatory Response Syndrome/microbiology , Time Factors , Tumor Necrosis Factor-alpha/analysis
7.
Mol Biol Cell ; 12(12): 3987-99, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11739795

ABSTRACT

In normal cells, activation of cyclin-dependent kinases (cdks) requires binding to a cyclin and phosphorylation by the cdk-activating kinase (CAK). The Kaposi's sarcoma-associated herpesvirus encodes a protein with similarity to D-type cyclins. This KSHV-cyclin activates CDK6, alters its substrate specificity, and renders CDK6 insensitive to inhibition by the cdk inhibitor p16(INK4a). Here we investigate the regulation of the CDK6/KSHV-cyclin kinase with the use of purified proteins and a cell-based assay. We find that KSHV-cyclin can activate CDK6 independent of phosphorylation by CAK in vitro. In addition, CAK phosphorylation decreased the p16(INK4a) sensitivity of CDK6/KSHV-cyclin complexes. In cells, expression of CDK6 or to a lesser degree of a nonphosphorylatable CDK6(T177A) together with KSHV-cyclin induced apoptosis, indicating that CDK6 activation by KSHV-cyclin can proceed in the absence of phosphorylation by CAK in vivo. Coexpression of p16 partially protected cells from cell death. p16 and KSHV-cyclin can form a ternary complex with CDK6 that can be detected by binding assays as well as by conformational changes in CDK6. The Kaposi's sarcoma-associated herpesvirus has adopted a clever strategy to render cell cycle progression independent of mitogenic signals, cdk inhibition, or phosphorylation by CAK.


Subject(s)
Apoptosis/drug effects , Cyclin-Dependent Kinases , Cyclins/pharmacology , Herpesvirus 8, Human , Protein Serine-Threonine Kinases/metabolism , Viral Proteins/pharmacology , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/pharmacology , Enzyme Activation/drug effects , Fluorescent Antibody Technique , Humans , Phosphorylation/drug effects , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Tumor Cells, Cultured , Cyclin-Dependent Kinase-Activating Kinase
8.
J Biol Chem ; 275(42): 32578-84, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-10931829

ABSTRACT

Cyclin-dependent kinases (CDKs) that control cell cycle progression are regulated in many ways, including activating phosphorylation of a conserved threonine residue. This essential phosphorylation is carried out by the CDK-activating kinase (CAK). Here we examine the effects of replacing this threonine residue in human CDK2 by serine. We found that cyclin A bound equally well to wild-type CDK2 (CDK2(Thr-160)) or to the mutant CDK2 (CDK2(Ser-160)). In the absence of activating phosphorylation, CDK2(Ser-160)-cyclin A complexes were more active than wild-type CDK2(Thr-160)-cyclin A complexes. In contrast, following activating phosphorylation, CDK2(Ser-160)-cyclin A complexes were less active than phosphorylated CDK2(Thr-160)-cyclin A complexes, reflecting a much smaller effect of activating phosphorylation on CDK2(Ser-160). The kinetic parameters for phosphorylating histone H1 were similar for mutant and wild-type CDK2, ruling out a general defect in catalytic activity. Interestingly, the CDK2(Ser-160) mutant was selectively defective in phosphorylating a peptide derived from the C-terminal domain of RNA polymerase II. CDK2(Ser-160) was efficiently phosphorylated by CAKs, both human p40(MO15)(CDK7)-cyclin H and budding yeast Cak1p. In fact, the k(cat) values for phosphorylation of CDK2(Ser-160) were significantly higher than for phosphorylation of CDK2(Thr-160), indicating that CDK2(Ser-160) is actually phosphorylated more efficiently than wild-type CDK2. In contrast, dephosphorylation proceeded more slowly with CDK2(Ser-160) than with wild-type CDK2, either in HeLa cell extract or by purified PP2Cbeta. Combined with the more efficient phosphorylation of CDK2(Ser-160) by CAK, we suggest that one reason for the conservation of threonine as the site of activating phosphorylation may be to favor unphosphorylated CDKs following the degradation of cyclins.


Subject(s)
CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Serine , Threonine , Amino Acid Substitution , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/genetics , Humans , Kinetics , Mutagenesis, Site-Directed , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity , Cyclin-Dependent Kinase-Activating Kinase
9.
J Biol Chem ; 275(43): 33267-71, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-10934199

ABSTRACT

Cak1p, the Cyclin-dependent kinase-activating kinase from budding yeast, is an unusual protein kinase that lacks many of the highly conserved motifs observed among members of the protein kinase superfamily. Cak1p phosphorylates and activates Cdc28p, the major cyclin-dependent kinase (CDK) in yeast, and is thereby required for passage through the yeast cell cycle. In this paper, we explore the kinetics of CDK phosphorylation by Cak1p, and we examine the role of the catalytic step in the reaction mechanism. Cak1p proceeds by a sequential reaction mechanism, binding to both ATP and CDK2 with reasonable affinities, exhibiting K(d) values of 7.2 and 0.6 microm, respectively. Interestingly, these values are approximately the same as the K(M) values, indicating that the binding of substrates is fast with respect to catalysis and that the most likely reaction mechanism is rapid equilibrium random. Cak1p is a slow enzyme, with a catalytic rate of only 4.3 min(-)(1). The absence of a burst phase indicates that product release is not rate-limiting. This result, and a solvent isotope effect, suggests that a catalytic step is rate-limiting.


Subject(s)
CDC2-CDC28 Kinases , Protein Serine-Threonine Kinases/metabolism , Saccharomycetales/enzymology , Adenosine Triphosphate/pharmacology , Catalysis , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Enzyme Stability , Kinetics , Phosphorylation , Cyclin-Dependent Kinase-Activating Kinase
10.
J Biol Chem ; 275(44): 34744-9, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10934208

ABSTRACT

We previously reported that the activating phosphorylation on cyclin-dependent kinases in yeast (Cdc28p) and in humans (Cdk2) is removed by type 2C protein phosphatases. In this study, we characterize this PP2C-like activity in HeLa cell extract and determine that it is due to PP2C beta 2, a novel PP2C beta isoform, and to PP2C alpha. PP2C alpha and PP2C beta 2 co-purified with Mg(2+)-dependent Cdk2/Cdk6 phosphatase activity in DEAE-Sepharose, Superdex-200, and Mono Q chromatographies. Moreover, purified recombinant PP2C alpha and PP2C beta 2 proteins efficiently dephosphorylated monomeric Cdk2/Cdk6 in vitro. The dephosphorylation of Cdk2 and Cdk6 by PP2C isoforms was inhibited by the binding of cyclins. We found that the PP2C-like activity in HeLa cell extract, partially purified HeLa PP2C alpha and PP2C beta 2 isoforms, and the recombinant PP2Cs exhibited a comparable substrate preference for a phosphothreonine containing substrate, consistent with the conservation of threonine residues at the site of activating phosphorylation in CDKs.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Isoenzymes/metabolism , Phosphoprotein Phosphatases/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , Chromatography, Ion Exchange , Cyclins/metabolism , HeLa Cells , Humans , Isoenzymes/chemistry , Isoenzymes/isolation & purification , Mice , Molecular Sequence Data , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/isolation & purification , Phosphorylation , Protein Phosphatase 2 , Protein Phosphatase 2C , Rats , Sequence Homology, Amino Acid , Substrate Specificity
11.
Eur J Biochem ; 267(13): 4213-21, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10866826

ABSTRACT

The cdk-activating kinase (CAK) activates cyclin-dependent kinases (cdks) that control cell-cycle progression by phosphorylating a threonine residue conserved in cdks. CAK from humans contains p40MO15 (cdk7), cyclin H and MAT1, which are also subunits of transcription factor IIH where they phosphorylate the C-terminal domain of the large subunit of RNA polymerase II. In contrast, budding yeast Cak1p is a monomeric enzyme without C-terminal domain kinase activity. Here, we analyze CAK activities in HeLa cells using cdk2-affinity chromatography. In addition to MO15, a second CAK activity was detected that runs on gel filtration at 30-40 kDa. This activity phosphorylated and activated cdk2 and cdk6. Furthermore, this 'small CAK' activity resembled Cak1p rather than MO15 in terms of substrate specificity, reactivity to antibodies against MO15 and Cak1p, and sensitivity to 5'-fluorosulfonylbenzoyladenosine, an irreversible inhibitory ATP analog. Our findings suggest the presence of at least two different CAK activities in human cells.


Subject(s)
CDC2-CDC28 Kinases , Protein Serine-Threonine Kinases/metabolism , Chromatography, Affinity , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , HeLa Cells , Humans , Phosphorylation , Substrate Specificity , Cyclin-Dependent Kinase-Activating Kinase
12.
Mol Biol Cell ; 11(5): 1597-609, 2000 May.
Article in English | MEDLINE | ID: mdl-10793138

ABSTRACT

Eukaryotic cell cycle progression is controlled by a family of protein kinases known as cyclin-dependent kinases (Cdks). Two steps are essential for Cdk activation: binding of a cyclin and phosphorylation on a conserved threonine residue by the Cdk-activating kinase (CAK). We have studied the interplay between these regulatory mechanisms during the activation of the major Saccharomyces cerevisiae Cdk, Cdc28p. We found that the majority of Cdc28p was phosphorylated on its activating threonine (Thr-169) throughout the cell cycle. The extent of Thr-169 phosphorylation was similar for monomeric Cdc28p and Cdc28p bound to cyclin. By varying the order of the addition of cyclin and Cak1p, we determined that Cdc28p was activated most efficiently when it was phosphorylated before cyclin binding. Furthermore, we found that a Cdc28p(T169A) mutant, which cannot be phosphorylated, bound cyclin less well than wild-type Cdc28p in vivo. These results suggest that unphosphorylated Cdc28p may be unable to bind tightly to cyclin. We propose that Cdc28p is normally phosphorylated by Cak1p before it binds cyclin. This activation pathway contrasts with that in higher eukaryotes, in which cyclin binding appears to precede activating phosphorylation.


Subject(s)
CDC28 Protein Kinase, S cerevisiae/metabolism , Cyclin-Dependent Kinases , Cyclins/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Antibody Specificity , CDC28 Protein Kinase, S cerevisiae/genetics , CDC28 Protein Kinase, S cerevisiae/immunology , Cell Cycle , Cyclin A/metabolism , Cyclin B/genetics , Cyclin B/metabolism , Enzyme Activation , Epitopes , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Threonine/immunology , Threonine/metabolism , Cyclin-Dependent Kinase-Activating Kinase
13.
Genes Dev ; 13(22): 2946-57, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10580002

ABSTRACT

Activating phosphorylation of cyclin-dependent protein kinases (CDKs) is necessary for their kinase activity and cell cycle progression. This phosphorylation is carried out by the Cdk-activating kinase (CAK); in contrast, little is known about the corresponding protein phosphatase. We show that type 2C protein phosphatases (PP2Cs) are responsible for this dephosphorylation of Cdc28p, the major budding yeast CDK. Two yeast PP2Cs, Ptc2p and Ptc3p, display Cdc28p phosphatase activity in vitro and in vivo, and account for approximately 90% of Cdc28p phosphatase activity in yeast extracts. Overexpression of PTC2 or PTC3 results in synthetic lethality in strains temperature-sensitive for yeast CAK1, and disruptions of PTC2 and PTC3 suppress the growth defect of a cak1 mutant. Furthermore, PP2C-like enzymes are the predominant phosphatases toward human Cdk2 in HeLa cell extracts, indicating that the substrate specificity of PP2Cs toward CDKs is evolutionarily conserved.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Fungal Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , CDC28 Protein Kinase, S cerevisiae/metabolism , Cell Cycle , Enzyme Activation , Gene Expression Regulation, Fungal , Humans , Phosphorylation , Phosphothreonine/metabolism , Protein Phosphatase 2 , Protein Phosphatase 2C , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/metabolism , Species Specificity , Cyclin-Dependent Kinase-Activating Kinase
14.
Proc Natl Acad Sci U S A ; 96(26): 14961-6, 1999 Dec 21.
Article in English | MEDLINE | ID: mdl-10611320

ABSTRACT

Transforming growth factor beta (TGF-beta)-mediated G(1) arrest previously has been shown to specifically target inactivation of cyclin D:cyclin-dependent kinase (Cdk) 4/6 complexes. We report here that TGF-beta-treated human HepG2 hepatocellular carcinoma cells arrest in G(1), but retain continued cyclin D:Cdk4/6 activity and active, hypophosphorylated retinoblastoma tumor suppressor protein. Consistent with this observation, TGF-beta-treated cells failed to induce p15(INK4b), down-regulate CDC25A, or increase levels of p21(CIP1), p27(KIP1), and p57(KIP2). However, TGF-beta treatment resulted in the specific inactivation of cyclin E:Cdk2 complexes caused by absence of the activating Thr(160) phosphorylation on Cdk2. Whole-cell lysates from TGF-beta-treated cells showed inhibition of Cdk2 Thr(160) Cdk activating kinase (CAK) activity; however, cyclin H:Cdk7 activity, a previously assumed mammalian CAK, was not altered. Saccharomyces cerevisiae contains a genetically and biochemically proven CAK gene, CAK1, that encodes a monomeric 44-kDa Cak1p protein unrelated to Cdk7. Anti-Cak1p antibodies cross-reacted with a 45-kDa human protein with CAK activity that was specifically down-regulated in response to TGF-beta treatment. Taken together, these observations demonstrate that TGF-beta signaling mediates a G(1) arrest in HepG2 cells by targeting Cdk2 CAK and suggests the presence of at least two mammalian CAKs: one specific for Cdk2 and one for Cdk4/6.


Subject(s)
CDC2-CDC28 Kinases , Cyclin E/metabolism , Cyclin-Dependent Kinases/metabolism , G1 Phase/drug effects , Protein Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/pharmacology , Cyclin H , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Down-Regulation , Enzyme Activation , Humans , Models, Biological , Phosphorylation , Threonine/metabolism , Tumor Cells, Cultured , Cyclin-Dependent Kinase-Activating Kinase
15.
Mol Cell Biol ; 19(7): 4774-87, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10373527

ABSTRACT

Cyclin-dependent kinase (CDK)-activating kinases (CAKs) carry out essential activating phosphorylations of CDKs such as Cdc2 and Cdk2. The catalytic subunit of mammalian CAK, MO15/Cdk7, also functions as a subunit of the general transcription factor TFIIH. However, these functions are split in budding yeast, where Kin28p functions as the kinase subunit of TFIIH and Cak1p functions as a CAK. We show that Kin28p, which is itself a CDK, also contains a site of activating phosphorylation on Thr-162. The kinase activity of a T162A mutant of Kin28p is reduced by approximately 75 to 80% compared to that of wild-type Kin28p. Moreover, cells containing kin28(T162A) and a conditional allele of TFB3 (the ortholog of the mammalian MAT1 protein, an assembly factor for MO15 and cyclin H) are severely compromised and display a significant further reduction in Kin28p activity. This finding provides in vivo support for the previous biochemical observation that MO15-cyclin H complexes can be activated either by activating phosphorylation of MO15 or by binding to MAT1. Finally, we show that Kin28p is no longer phosphorylated on Thr-162 following inactivation of Cak1p in vivo, that Cak1p can phosphorylate Kin28p on Thr-162 in vitro, and that this phosphorylation stimulates the CTD kinase activity of Kin28p. Thus, Kin28p joins Cdc28p, the major cell cycle Cdk in budding yeast, as a physiological Cak1p substrate. These findings indicate that although MO15 and Cak1p constitute different forms of CAK, both control the cell cycle and the phosphorylation of the C-terminal domain of the large subunit of RNA polymerase II by TFIIH.


Subject(s)
Cyclin-Dependent Kinases , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/enzymology , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Transcription Factors, TFII , Transcription Factors/metabolism , Animals , Cell Cycle , Enzyme Activation , Phosphorylation , Point Mutation , Protein Serine-Threonine Kinases/genetics , Rabbits , Saccharomyces cerevisiae/genetics , Threonine/metabolism , Transcription Factor TFIIH , Cyclin-Dependent Kinase-Activating Kinase
16.
Cell Mol Life Sci ; 55(2): 284-96, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10188587

ABSTRACT

Cell cycle progression is regulated by cyclin-dependent kinases (cdks). The activity of cdks is tightly controlled by several mechanisms, including binding of subunits to cdks (cyclins and inhibitors), and phosphorylation events. This review focuses on the activating phosphorylation of cdks by an enzyme termed cdk-activating kinase (CAK). Two classes of CAKs have been identified: monomeric Cak1p from budding yeast and the p40MO15 (cdk7)/cyclin H/MAT1 complex from vertebrates. Cak1p is the physiological CAK in budding yeast and localizes to the cytoplasm. p40MO15(cdk7)/cyclin H/MAT1 localizes to the nucleus, is a subunit of the general transcription factor IIH and activates cdks as well as phosphorylates several components of the transcriptional machinery. Functions, substrate specificities, regulation, localization, effects on cdk structure and involvement in transcription are compared for Cak1p and p40MO15(cdk7).


Subject(s)
Cyclin-Dependent Kinases , Protein Serine-Threonine Kinases/physiology , Animals , Humans , Mammals , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Yeasts/enzymology , Cyclin-Dependent Kinase-Activating Kinase
17.
J Biol Chem ; 274(4): 1949-56, 1999 Jan 22.
Article in English | MEDLINE | ID: mdl-9890950

ABSTRACT

Cak1p is an essential protein kinase that phosphorylates and thereby activates the major cyclin-dependent kinase in budding yeast, Cdc28p. The sequence of Cak1p differs from other members of the protein kinase superfamily in several conserved regions. Cak1p lacks the highly conserved glycine loop motif (GXGXXG) that is found in the nucleotide binding fold of virtually all protein kinases and also lacks a number of conserved amino acids found at sites throughout the protein kinase core sequence. We have used kinetic and mutagenic analyses to investigate whether these sequence differences affect the nucleotide-binding properties of Cak1p. Although Cak1p differs dramatically from other protein kinases, it binds ATP with a reasonable affinity, with a KM of 4.8 microM. Mutations of the putative invariant lysine in Cak1p (Lys-31), homologous to a residue required for activity in virtually all protein kinases and that interacts with the ATP phosphates, moderately reduced the ability of Cak1p to bind ATP but did not dramatically affect the catalytic rate of the kinase. Similarly, Cak1p is insensitive to the ATP analog 5'-fluorosulfonylbenzoyladenosine, which inhibits most protein kinases through covalent modification of the invariant lysine. We found that Cak1p is tolerant of mutations within its glycine loop region. Remarkably, Cak1p remains functional even following truncation of its first 31 amino acids, including the glycine loop region and the invariant lysine. We conclude that the Cak1p nucleotide-binding pocket differs significantly from those of most other protein kinases and therefore might provide a specific target for an inhibitory drug.


Subject(s)
Adenosine Triphosphate/metabolism , Cyclin-Dependent Kinases , Protein Serine-Threonine Kinases/metabolism , Schizosaccharomyces/enzymology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/analogs & derivatives , Amino Acid Sequence , Base Sequence , DNA Primers , Kinetics , Molecular Sequence Data , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Sequence Homology, Amino Acid , Cyclin-Dependent Kinase-Activating Kinase
18.
J Cell Sci ; 111 ( Pt 24): 3585-96, 1998 Dec 18.
Article in English | MEDLINE | ID: mdl-9819350

ABSTRACT

Eukaryotic cell cycles are controlled by the activities of cyclin-dependent kinases (cdks). The major cdk in budding yeast, Saccharomyces cerevisiae, is Cdc28p. Activation of Cdc28p requires phosphorylation on threonine 169 and binding to a cyclin. Thr-169 is phosphorylated by the cdk-activating kinase (CAK), Cak1p, which was recently identified as the physiological CAK in budding yeast. Here we present our further characterization of yeast Cak1p. We have found that Cak1p is dispersed throughout the cell as shown by immunofluorescence; biochemical subcellular fractionation confirmed that most of the Cak1p is found in the cytoplasm. Cak1p is a monomeric enzyme in crude yeast lysates. Mutagenesis of potential sites of activating phosphorylation had little effect on the activity of Cak1p in vitro or in vivo. Furthermore, Cak1p contains no posttranslational modifications detectable by two-dimensional isoelectric focusing. We found that Cak1p is a stable protein during exponential growth but that its expression decreases considerably when cells enter stationary phase. In contrast, Cak1p levels oscillate dramatically during meiosis, reflecting regulation at both the transcriptional and post-translational level. The localization and regulation of Cak1p are in contrast to those of the known vertebrate CAK, p40(MO15).


Subject(s)
Cyclin-Dependent Kinases , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/enzymology , Amino Acid Sequence , CDC28 Protein Kinase, S cerevisiae/metabolism , Cytoplasm/enzymology , Fluorescent Antibody Technique , Gene Expression Regulation, Fungal , Isoelectric Focusing , Meiosis/physiology , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Saccharomyces cerevisiae/physiology , Subcellular Fractions/enzymology , Cyclin-Dependent Kinase-Activating Kinase
19.
Mol Biol Cell ; 9(9): 2545-60, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9725911

ABSTRACT

Cell cycle progression is controlled by the sequential functions of cyclin-dependent kinases (cdks). Cdk activation requires phosphorylation of a key residue (on sites equivalent to Thr-160 in human cdk2) carried out by the cdk-activating kinase (CAK). Human CAK has been identified as a p40(MO15)/cyclin H/MAT1 complex that also functions as part of transcription factor IIH (TFIIH) where it phosphorylates multiple transcriptional components including the C-terminal domain (CTD) of the large subunit of RNA polymerase II. In contrast, CAK from budding yeast consists of a single polypeptide (Cak1p), is not a component of TFIIH, and lacks CTD kinase activity. Here we report that Cak1p and p40(MO15) have strikingly different substrate specificities. Cak1p preferentially phosphorylated monomeric cdks, whereas p40(MO15) preferentially phosphorylated cdk/cyclin complexes. Furthermore, p40(MO15) only phosphorylated cdk6 bound to cyclin D3, whereas Cak1p recognized monomeric cdk6 and cdk6 bound to cyclin D1, D2, or D3. We also found that cdk inhibitors, including p21(CIP1), p27(KIP1), p57(KIP2), p16(INK4a), and p18(INK4c), could block phosphorylation by p40(MO15) but not phosphorylation by Cak1p. Our results demonstrate that although both Cak1p and p40(MO15) activate cdks by phosphorylating the same residue, the structural mechanisms underlying the enzyme-substrate recognition differ greatly. Structural and physiological implications of these findings will be discussed.


Subject(s)
CDC2-CDC28 Kinases , Protein Serine-Threonine Kinases/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Enzyme Activation , Humans , Phosphorylation , Substrate Specificity , Yeasts/enzymology , Cyclin-Dependent Kinase-Activating Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...