Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
NPJ Syst Biol Appl ; 6(1): 8, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32245958

ABSTRACT

Some biological networks exhibit oscillations in their components to convert stimuli to time-dependent responses. The eukaryotic cell cycle is such a case, being governed by waves of cyclin-dependent kinase (cyclin/Cdk) activities that rise and fall with specific timing and guarantee its timely occurrence. Disruption of cyclin/Cdk oscillations could result in dysfunction through reduced cell division. Therefore, it is of interest to capture properties of network designs that exhibit robust oscillations. Here we show that a minimal yeast cell cycle network is able to oscillate autonomously, and that cyclin/Cdk-mediated positive feedback loops (PFLs) and Clb3-centered regulations sustain cyclin/Cdk oscillations, in known and hypothetical network designs. We propose that Clb3-mediated coordination of cyclin/Cdk waves reconciles checkpoint and oscillatory cell cycle models. Considering the evolutionary conservation of the cyclin/Cdk network across eukaryotes, we hypothesize that functional ("healthy") phenotypes require the capacity to oscillate autonomously whereas dysfunctional (potentially "diseased") phenotypes may lack this capacity.


Subject(s)
Biological Clocks/physiology , Cyclin B/metabolism , Cyclins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Cell Cycle/physiology , Cell Cycle Checkpoints/genetics , Cell Division , Cyclin B/genetics , Cyclin B/physiology , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Models, Biological , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Systems Biology/methods
2.
J Cell Biol ; 218(5): 1553-1563, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30770433

ABSTRACT

Meiosis with a single round of DNA replication and two successive rounds of chromosome segregation requires specific cyclins associated with cyclin-dependent kinases (CDKs) to ensure its fidelity. But how cyclins control the distinctive meiosis is still largely unknown. In this study, we explored the role of cyclin B3 in female meiosis by generating Ccnb3 mutant mice via CRISPR/Cas9. Ccnb3 mutant oocytes characteristically arrested at metaphase I (MetI) with normal spindle assembly and lacked enough anaphase-promoting complex/cyclosome (APC/C) activity, which is spindle assembly checkpoint (SAC) independent, to initiate anaphase I (AnaI). Securin siRNA or CDK1 inhibitor supplements rescued the MetI arrest. Furthermore, CCNB3 directly interacts with CDK1 to exert kinase function. Besides, the MetI arrest oocytes had normal development after intracytoplasmic sperm injection (ICSI) or parthenogenetic activation (PA), along with releasing the sister chromatids, which implies that Ccnb3 exclusively functioned in meiosis I, rather than meiosis II. Our study sheds light on the specific cell cycle control of cyclins in meiosis.


Subject(s)
Anaphase/physiology , Chromosome Segregation , Cyclin B/physiology , Kinetochores/physiology , Meiosis/physiology , Metaphase/physiology , Oocytes/physiology , Anaphase-Promoting Complex-Cyclosome/metabolism , Animals , CDC2 Protein Kinase/metabolism , Embryonic Development , Female , M Phase Cell Cycle Checkpoints , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Oocytes/cytology
3.
J Cell Sci ; 130(16): 2722-2735, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28705837

ABSTRACT

Depletion of cyb-1, a major B-type cyclin expressed during Caenorhabditis elegans spermatogenesis, causes a meiotic division arrest in diakinesis-stage spermatocytes with multiple and mispositioned centrosomes. Association of the two nuclear membrane proteins SUN-1 and ZYG-12 is essential for centrosome-nuclear envelope attachment. We found that depletion of sun-1 causes centrosome defects similar to those caused by cyb-1 depletion in diakinesis-stage spermatocytes. In addition, Ser8 and Ser43 residues in SUN-1 are dephosphorylated in cyb-1-depleted diakinesis-stage spermatocytes. Nevertheless, dephosphorylation of these residues was not sufficient to reproduce the cyb-1-related centrosome defects. We then found that the ZYG-12::GFP signal in the nuclear envelope was significantly reduced in the cyb-1-depleted diakinesis-stage spermatocytes. However, only mispositioned but not multiplied centrosomes were observed in zyg-12 mutant diakinesis-stage spermatocytes, suggesting that zyg-12 is not involved in the centrosome duplication at this stage. Our results suggest that CYB-1 functions to maintain proper positioning of centrosomes during spermatogenesis by regulating phosphorylation of SUN-1, which is possibly crucial for the association between SUN-1 and ZYG-12. This phosphorylation of SUN-1 may also regulate centrosome duplication independently of ZYG-12.


Subject(s)
Caenorhabditis elegans/physiology , Centrosome/metabolism , Cyclin B/physiology , Spermatocytes/physiology , Spermatogenesis/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cyclin B/genetics , Male , Meiosis/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Spermatocytes/metabolism
5.
Cell Cycle ; 14(16): 2648-54, 2015.
Article in English | MEDLINE | ID: mdl-26125114

ABSTRACT

Cyclin B3 is a relatively new member of the cyclin family whose functions are little known. We found that depletion of cyclin B3 inhibited metaphase-anaphase transition as indicated by a well-sustained MI spindle and cyclin B1 expression in meiotic oocytes after extended culture. This effect was independent of spindle assembly checkpoint activity, since both Bub3 and BubR1 signals were not observed at kinetochores in MI-arrested cells. The metaphase I arrest was not rescued by either Mad2 knockdown or cdc20 overexpression, but it was rescued by securin RNAi. We conclude that cyclin B3 controls the metaphase-anaphase transition by activating APC/C(cdc20) in meiotic oocytes, a process that does not rely on SAC activity.


Subject(s)
Anaphase , Cyclin B/physiology , Oocytes/physiology , Animals , Cells, Cultured , Female , M Phase Cell Cycle Checkpoints , Meiosis , Mice, Inbred ICR
6.
Dev Biol ; 396(1): 67-80, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25264619

ABSTRACT

CDC6 is essential for S-phase to initiate DNA replication. It also regulates M-phase exit by inhibiting the activity of the major M-phase protein kinase CDK1. Here we show that addition of recombinant CDC6 to Xenopus embryo cycling extract delays the M-phase entry and inhibits CDK1 during the whole M-phase. Down regulation of endogenous CDC6 accelerates the M-phase entry, abolishes the initial slow and progressive phase of histone H1 kinase activation and increases the level of CDK1 activity during the M-phase. All these effects are fully rescued by the addition of recombinant CDC6 to the extracts. Diminution of CDC6 level in mouse zygotes by two different methods results in accelerated entry into the first cell division showing physiological relevance of CDC6 in intact cells. Thus, CDC6 behaves as CDK1 inhibitor regulating not only the M-phase exit, but also the M-phase entry and progression via limiting the level of CDK1 activity. We propose a novel mechanism of M-phase entry controlled by CDC6 and counterbalancing cyclin B-mediated CDK1 activation. Thus, CDK1 activation proceeds with concomitant inhibition by CDC6, which tunes the timing of the M-phase entry during the embryonic cell cycle.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cell Division , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Developmental , Nuclear Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cell Cycle/genetics , Cell-Free System , Cyclin B/physiology , DNA Replication , Enzyme Activation , Female , Glutathione Transferase/metabolism , Mice , Mitosis , Phosphorylation , Protein Kinases/metabolism , Recombinant Proteins/metabolism , Time Factors , Xenopus laevis
7.
Curr Biol ; 24(6): 646-51, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24583014

ABSTRACT

Chromosome attachment to the mitotic spindle in early mitosis is guarded by an Aurora B kinase-dependent error correction mechanism [1, 2] and by the spindle assembly checkpoint (SAC), which delays cell-cycle progression in response to errors in chromosome attachment [3, 4]. The abrupt loss of sister chromatid cohesion at anaphase creates a type of chromosome attachment that in early mitosis would be recognized as erroneous, would elicit Aurora B-dependent destabilization of kinetochore-microtubule attachment, and would activate the checkpoint [5, 6]. However, in anaphase, none of these responses occurs, which is vital to ensure progression through anaphase and faithful chromosome segregation. The difference has been attributed to the drop in CDK1/cyclin B activity that accompanies anaphase and causes Aurora B translocation away from centromeres [7-12] and to the inactivation of the checkpoint by the time of anaphase [10, 11, 13, 14]. Here, we show that checkpoint inactivation may not be crucial because checkpoint activation by anaphase chromosomes is too slow to take effect on the timescale during which anaphase is executed. In addition, we observe that checkpoint activation can still occur for a considerable time after the anaphase-promoting complex/cyclosome (APC/C) becomes active, raising the question whether the checkpoint is indeed completely inactivated by the time of anaphase under physiologic conditions.


Subject(s)
Anaphase/physiology , M Phase Cell Cycle Checkpoints/physiology , Aurora Kinase B/physiology , Chromatids/physiology , Cyclin B/physiology , Kinetics , Kinetochores/physiology , Saccharomyces , Securin/physiology , Separase/physiology , Spindle Apparatus/physiology
8.
Curr Biol ; 24(6): 630-7, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24583015

ABSTRACT

Activation of anaphase-promoting complex/cyclosome (APC/C(Cdc20)) by Cdc20 is delayed by the spindle assembly checkpoint (SAC). When all kinetochores come under tension, the SAC is turned off and APC/C(Cdc20) degrades cyclin B and securin, which activates separase [1]. The latter then cleaves cohesin holding sister chromatids together [2]. Because cohesin cleavage also destroys the tension responsible for turning off the SAC, cells must possess a mechanism to prevent SAC reactivation during anaphase, which could be conferred by a dependence of the SAC on Cdk1 [3-5]. To test this, we analyzed mouse oocytes and embryos expressing nondegradable cyclin B together with a Cdk1-resistant form of separase. After biorientation and SAC inactivation, APC/C(Cdc20) activates separase but the resulting loss of (some) cohesion is accompanied by SAC reactivation and APC/C(Cdc20) inhibition, which aborts the process of further securin degradation. Cyclin B is therefore the only APC/C(Cdc20) substrate whose degradation at the onset of anaphase is necessary to prevent SAC reactivation. The mutual activation of tension sensitive SAC and Cdk1 creates a bistable system that ensures complete activation of separase and total downregulation of Cdk1 when all chromosomes have bioriented.


Subject(s)
Anaphase/physiology , CDC2 Protein Kinase/physiology , M Phase Cell Cycle Checkpoints/physiology , Animals , Cdc20 Proteins/physiology , Chromatids/physiology , Cyclin B/physiology , Female , Male , Mice , Mice, Knockout , Nondisjunction, Genetic/physiology , Oocytes/physiology , Phosphorylation
9.
Curr Biol ; 24(6): 638-45, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24583019

ABSTRACT

Two mechanisms safeguard the bipolar attachment of chromosomes in mitosis. A correction mechanism destabilizes erroneous attachments that do not generate tension across sister kinetochores [1]. In response to unattached kinetochores, the mitotic checkpoint delays anaphase onset by inhibiting the anaphase-promoting complex/cyclosome (APC/C(Cdc20)) [2]. Upon satisfaction of both pathways, the APC/C(Cdc20) elicits the degradation of securin and cyclin B [3]. This liberates separase triggering sister chromatid disjunction and inactivates cyclin-dependent kinase 1 (Cdk1) causing mitotic exit. How eukaryotic cells avoid the engagement of attachment monitoring mechanisms when sister chromatids split and tension is lost at anaphase is poorly understood [4]. Here we show that Cdk1 inactivation disables mitotic checkpoint surveillance at anaphase onset in human cells. Preventing cyclin B1 proteolysis at the time of sister chromatid disjunction destabilizes kinetochore-microtubule attachments and triggers the engagement of the mitotic checkpoint. As a consequence, mitotic checkpoint proteins accumulate at anaphase kinetochores, the APC/C(Cdc20) is inhibited, and securin reaccumulates. Conversely, acute pharmacological inhibition of Cdk1 abrogates the engagement and maintenance of the mitotic checkpoint upon microtubule depolymerization. We propose that the simultaneous destruction of securin and cyclin B elicited by the APC/C(Cdc20) couples chromosome segregation to the dissolution of attachment monitoring mechanisms during mitotic exit.


Subject(s)
Anaphase/physiology , CDC2 Protein Kinase/physiology , Kinetochores/physiology , M Phase Cell Cycle Checkpoints/physiology , Chromatids/physiology , Cyclin B/physiology , Cyclin B1/physiology , HeLa Cells , Humans , Nondisjunction, Genetic/physiology , Separase/physiology
10.
Nat Struct Mol Biol ; 20(12): 1415-24, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24186061

ABSTRACT

The order and timing of cell-cycle events is controlled by changing substrate specificity and different activity thresholds of cyclin-dependent kinases (CDKs). However, it is not understood how a single protein kinase can trigger hundreds of switches in a sufficiently time-resolved fashion. We show that cyclin-Cdk1-Cks1-dependent phosphorylation of multisite targets in Saccharomyces cerevisiae is controlled by key substrate parameters including distances between phosphorylation sites, distribution of serines and threonines as phosphoacceptors and positioning of cyclin-docking motifs. The component mediating the key interactions in this process is Cks1, the phosphoadaptor subunit of the cyclin-Cdk1-Cks1 complex. We propose that variation of these parameters within networks of phosphorylation sites in different targets provides a wide range of possibilities for differential amplification of Cdk1 signals, thus providing a mechanism to generate a wide range of thresholds in the cell cycle.


Subject(s)
CDC2 Protein Kinase/physiology , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/physiology , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Cyclin B/metabolism , Cyclin B/physiology , Cyclins/metabolism , Cyclins/physiology , Phosphorylation , Phosphoserine/chemistry , Phosphoserine/metabolism , Phosphothreonine/chemistry , Phosphothreonine/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Signal Transduction
11.
Biochem Pharmacol ; 83(2): 199-206, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22024133

ABSTRACT

Mcl-1 is one of the major anti-apoptotic members of the Bcl-2 family of apoptotic regulatory proteins. In this study we investigated the role of Mcl-1 in mitotic arrest-induced apoptosis. Vinblastine treatment of KB-3 cells initially resulted in a phosphatase-sensitive mobility shift in Mcl-1 and then subsequent loss of Mcl-1 protein expression which was prevented by MG132, suggesting that phosphorylation triggered proteosome-mediated degradation. Mcl-1 phosphorylation/degradation was a specific response to microtubule inhibition and did not occur in response to lethal concentrations of DNA damaging agents. Vinblastine treatment caused degradation of Mcl-1 in cells in which apoptosis was blocked by Bcl-xL overexpression, indicating that Mcl-1 degradation was not a consequence of apoptosis. A partial reversible phosphorylation of Mcl-1 was observed in synchronized cells traversing mitosis, whereas more extensive phosphorylation and subsequent degradation of Mcl-1 was observed if synchronized cells were treated with vinblastine. Mcl-1 phosphorylation closely paralleled cyclin B expression, and specific cyclin-dependent kinase (Cdk) inhibitors blocked vinblastine-induced Mcl-1 phosphorylation, its subsequent degradation, and improved cell viability after mitotic arrest. Co-immunoprecipitation studies indicated that Mcl-1 was complexed with Bak, but not Bax or Noxa, in untreated cells, and that Bak became activated in concert with loss of Mcl-1 expression. These results suggest that Cdk1/cyclin B plays a key role in mitotic arrest-induced apoptosis via Mcl-1 phosphorylation, promoting its degradation and subsequently releasing Bak from sequestration.


Subject(s)
Apoptosis/physiology , CDC2 Protein Kinase/physiology , Cyclin B/physiology , Mitosis/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Humans , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation/physiology , bcl-2-Associated X Protein/metabolism
12.
J Cell Biol ; 193(7): 1229-44, 2011 Jun 27.
Article in English | MEDLINE | ID: mdl-21690306

ABSTRACT

In animals, the female meiotic spindle is positioned at the egg cortex in a perpendicular orientation to facilitate the disposal of half of the chromosomes into a polar body. In Caenorhabditis elegans, the metaphase spindle lies parallel to the cortex, dynein is dispersed on the spindle, and the dynein activators ASPM-1 and LIN-5 are concentrated at spindle poles. Anaphase-promoting complex (APC) activation results in dynein accumulation at spindle poles and dynein-dependent rotation of one spindle pole to the cortex, resulting in perpendicular orientation. To test whether the APC initiates spindle rotation through cyclin B-CDK-1 inactivation, separase activation, or degradation of an unknown dynein inhibitor, CDK-1 was inhibited with purvalanol A in metaphase-I-arrested, APC-depleted embryos. CDK-1 inhibition resulted in the accumulation of dynein at spindle poles and dynein-dependent spindle rotation without chromosome separation. These results suggest that CDK-1 blocks rotation by inhibiting dynein association with microtubules and with LIN-5-ASPM-1 at meiotic spindle poles and that the APC promotes spindle rotation by inhibiting CDK-1.


Subject(s)
CDC2 Protein Kinase/physiology , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/genetics , Dyneins/physiology , Meiosis/physiology , Spindle Apparatus/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , CDC2 Protein Kinase/antagonists & inhibitors , Caenorhabditis elegans/cytology , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/metabolism , Cell Cycle Proteins/metabolism , Cyclin B/physiology , Cytoplasmic Dyneins/metabolism , Cytoplasmic Dyneins/physiology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Enzyme Inhibitors/pharmacology , Microtubules/metabolism , Purines/pharmacology , Spindle Apparatus/ultrastructure , Ubiquitin-Protein Ligase Complexes/physiology
13.
Mol Reprod Dev ; 78(10-11): 704-7, 2011.
Article in English | MEDLINE | ID: mdl-21714029

ABSTRACT

This short review updates the maturation-inducing hormonal signaling in starfish oocytes. In this system, the activation of cyclin B-Cdc2 kinase (Cdk1) that leads to meiotic resumption does not require new protein synthesis. The key intracellular mediator after hormonal stimulation by 1-methyladenine is the protein kinase Akt/PKB, which in turn directly downregulates Myt1 and upregulates Cdc25 toward the activation of cyclin B-Cdc2. Mitotic kinases including Aurora, Plk1 and Greatwall are activated downstream of cyclin B-Cdc2. The starfish oocyte thus provides a simple model system for the study of meiotic resumption.


Subject(s)
Maturation-Promoting Factor/pharmacology , Meiosis/drug effects , Oocytes/drug effects , Starfish/genetics , Starfish/metabolism , Animals , CDC2 Protein Kinase/metabolism , CDC2 Protein Kinase/physiology , Cyclin B/metabolism , Cyclin B/physiology , Female , Hormones/pharmacology , Meiosis/physiology , Models, Biological , Oocytes/metabolism , Oocytes/physiology , Signal Transduction/drug effects , Signal Transduction/genetics
14.
J Theor Biol ; 283(1): 103-12, 2011 Aug 21.
Article in English | MEDLINE | ID: mdl-21635899

ABSTRACT

The process of cell division in mammalian cells is orchestrated by cell-cycle-dependent oscillations of cyclin protein levels. Cyclin levels are controlled by redundant transcriptional, post-translational and degradation feedback loops. How each of these separate loops contributes to the regulation of the key cell cycle events and to the connection between the G1-S transition and the subsequent mitotic events is under investigation. Here, we present an integrated computational model of the mammalian cell cycle based on the sequential activation of cyclins. We validate the model against experimental data on liver cells (hepatocytes), which undergo one or two rounds of synchronous circadian-clock gated cell divisions during liver regeneration, after partial hepatectomy (PH). The model exhibits bandpass filter properties that allow the system to ignore strong but transient, or sustained but weak damages after PH. Bifurcation analysis of the model suggests two different threshold mechanisms for the progression of the cell through mitosis. These results are coherent with the notion that the mitotic exit in mammalian cells is bistable, and suggests that Cdc20 homologue 1 (Cdh1) is an important regulator of mitosis. Regulation by Cdh1 also explains the observed G2/M phase prolongation after hepatocyte growth factor (HGF) stimulation during S phase.


Subject(s)
Cell Cycle/physiology , Liver Regeneration/physiology , Mammals/physiology , Models, Biological , Animals , Cell Cycle/drug effects , Cell Division/physiology , Circadian Rhythm/physiology , Cyclin B/physiology , Cyclins/physiology , DNA/biosynthesis , Hepatectomy , Hepatocyte Growth Factor/pharmacology , Hepatocytes/cytology , Hepatocytes/physiology , Mitosis/physiology , Peptide Hydrolases/metabolism
16.
Mol Nutr Food Res ; 55(5): 749-60, 2011 May.
Article in English | MEDLINE | ID: mdl-21287681

ABSTRACT

SCOPE: Sterigmatocystin (ST), a mycotoxin commonly found in foodstuff and feedstuff, has been shown to be a carcinogenic mycotoxin in animal models. Many studies showed that the high level of ST contamination in grains might be related to the high incidence of gastric carcinoma in rural areas of China. However, up to now, the potential effects of ST on human gastric epithelium cells remain largely unknown. In this study, we explored the effects of ST on cell-cycle distribution and the regulatory mechanism in immortalized human gastric epithelium cells (GES-1). METHODS AND RESULTS: The effects of ST on the cell cycle distribution of GES-1 cells were determined with flow cytometric (FCM) analysis, Giemsa staining and immunofluorescence staining, while that on the expression of related gene-Cdc25C, Cdc2, CyclinB1 and the complex of CyclinB1-Cdc2 were studied with Western blot, reverse transcription polymerase chain reaction (RT-PCR) and immunoprecipitation assay respectively. We found that ST induced GES-1 cells arrested at G2 phase by regulating the expression of Cdc25C, Cdc2, CyclinB1 and the formation of CyclinB1-Cdc2 complex. Further study suggested JNK, ERK and PI3K/AKT/mTOR pathways to be involved in the process of G2 arrest induced by ST. The specific inhibitors of JNK and ERK reversed the role of ST, whereas that of PI3K/AKT/mTOR reinforced the effect of ST on cell-cycle distribution. CONCLUSION: This study demonstrates that JNK, ERK and PI3K/AKT/mTOR pathways participated in the G2 arrest induced by ST through the deregulation of CyclinB1, Cdc2 and Cdc25C. It may play some roles in the gastric carcinogenesis in ST exposure populations.


Subject(s)
G2 Phase/drug effects , Gastric Mucosa/drug effects , MAP Kinase Signaling System/physiology , Signal Transduction/physiology , Sterigmatocystin/toxicity , CDC2 Protein Kinase , Cells, Cultured , Cyclin B/physiology , Cyclin B1/physiology , Cyclin-Dependent Kinases , Gastric Mucosa/pathology , Humans , Phosphatidylinositol 3-Kinases/physiology , cdc25 Phosphatases/physiology
17.
Curr Genet ; 56(6): 479-93, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20697716

ABSTRACT

The Target of Rapamycin complex 1 (TORC1) is a central regulator of eukaryotic cell growth that is inhibited by the drug rapamycin. In the budding yeast Saccharomyces cerevisiae, translational defects associated with TORC1 inactivation inhibit cell cycle progression at an early stage in G1, but little is known about the possible roles for TORC1 later in the cell cycle. We investigated the rapamycin-hypersensitivity phenotype of cells lacking the S phase cyclin Clb5 (clb5Δ) as a basis for uncovering novel connections between TORC1 and the cell cycle regulatory machinery. Dosage suppression experiments suggested that the clb5Δ rapamycin hypersensitivity reflects a unique Clb5-associated cyclin-dependent kinase (CDK) function that cannot be performed by mitotic cyclins and that also involves motor proteins, particularly the kinesin-like protein Kip3. Synchronized cell experiments revealed rapamycin-induced defects in pre-anaphase spindle assembly and S phase progression that were more severe in clb5Δ than in wild-type cells but no apparent activation of Rad53-dependent checkpoint pathways. Some rapamycin-treated cells had aberrant spindle morphologies, but rapamycin did not cause gross defects in the microtubule cytoskeleton. We propose a model in which TORC1 and Clb5/CDK act coordinately to promote both spindle assembly via a pathway involving Kip3 and S phase progression.


Subject(s)
Cyclin B/physiology , DNA Replication/genetics , Multiprotein Complexes/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae , Spindle Apparatus/metabolism , TOR Serine-Threonine Kinases/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Cell Survival/drug effects , Cell Survival/genetics , Cyclin B/genetics , Cyclin B/metabolism , DNA Replication/drug effects , Drug Resistance/drug effects , Drug Resistance/genetics , Kinesins/genetics , Kinesins/metabolism , Kinesins/physiology , Multiprotein Complexes/metabolism , Organisms, Genetically Modified , Protein Multimerization/drug effects , Protein Multimerization/genetics , S Phase/drug effects , S Phase/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sirolimus/pharmacology , Spindle Apparatus/drug effects , Spindle Apparatus/genetics , TOR Serine-Threonine Kinases/metabolism
18.
FEBS J ; 276(21): 6063-73, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19788417

ABSTRACT

Cell death by the process of apoptosis plays important roles in development, tissue homeostasis, diseases and drug responses. The cysteine aspartyl protease caspase-9 plays a central role in the mitochondrial or intrinsic apoptotic pathway that is engaged in response to many apoptotic stimuli. Caspase-9 is activated in a large multimeric complex, the apoptosome, which is formed with apoptotic peptidase activating factor 1 (Apaf-1) in response to the release of cytochrome c from mitochondria. Once activated, caspase-9 cleaves and activates the effector caspases 3 and 7 to bring about apoptosis. This pathway is tightly regulated at multiple steps, including apoptosome formation and caspase-9 activation. Recent work has shown that caspase-9 is the direct target for regulatory phosphorylation by multiple protein kinases activated in response to extracellular growth/survival factors, osmotic stress or during mitosis. Here, we review these advances and discuss the possible roles of caspase-9 phosphorylation in the regulation of apoptosis during development and in pathological states, including cancer.


Subject(s)
Apoptosis , Autophagy , Caspase 9/metabolism , Amino Acid Sequence , Animals , CDC2 Protein Kinase/physiology , Caspase 9/chemistry , Caspase Inhibitors , Cyclin B/physiology , Cyclin B1 , DNA Damage , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Molecular Sequence Data , Phosphorylation , p38 Mitogen-Activated Protein Kinases/physiology
19.
J Biol Chem ; 284(42): 28775-82, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19687009

ABSTRACT

CLIP-170, the founding member of microtubule "plus ends tracking" proteins, is involved in many critical microtubule-related functions, including recruitment of dynactin to the microtubule plus ends and formation of kinetochore-microtubule attachments during metaphase. Although it has been reported that CLIP-170 is a phosphoprotein, neither have individual phosphorylation sites been identified nor have the associated kinases been extensively studied. Herein, we identify Cdc2 as a kinase that phosphorylates CLIP-170. We show that Cdc2 interacts with CLIP-170 mediating its phosphorylation on Thr(287) in vivo. Significantly, expression of CLIP-170 with a threonine 287 to alanine substitution (T287A) results in its mislocalization, accumulation of Plk1 and cyclin B, and block of the G2/M transition. Finally, we found that depletion of CLIP-170 leads to centrosome reduplication and that Cdc2 phosphorylation of CLIP-170 is required for the process. These results demonstrate that Cdc2-mediated phosphorylation of CLIP-170 is essential for the normal function of this protein during cell cycle progression.


Subject(s)
Centrosome/ultrastructure , Cyclin B/chemistry , Cyclin B/physiology , Microtubule-Associated Proteins/chemistry , Neoplasm Proteins/chemistry , Animals , CDC2 Protein Kinase , Cell Cycle , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinases , Humans , Microtubules/metabolism , Peptides/chemistry , Phenotype , Phosphorylation , Rats , Recombinant Proteins/chemistry , Tubulin/chemistry
20.
Cancer Sci ; 100(11): 2115-25, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19709076

ABSTRACT

The breast cancer susceptibility gene (BRCA2) is localized mainly in the nucleus where it plays an important role in DNA damage repair. Some BRCA2 protein is also present in the centrosome. Here, we demonstrate that BRCA2 interacts with plectin, a cytoskeletal cross-linker protein, and that this interaction controls the position of the centrosome. Phosphorylation of plectin by cyclin-dependent kinase 1/cyclin B (CDK1/CycB) kinase has been reported to abolish its cross-linking function during mitosis. Here, we induced phosphorylation of plectin in prepared fractions of HeLa cells by adding activated CDK1/CycB kinase. Consequently, there was significant dissociation of the centrosome from the nuclear membrane. Plectin has six homologous ankyrin-like repeat domains (termed PLEC M1-M6). Using a pull-down assay, we found that GST-PLEC M1 and a GST-C-terminal region fusion protein (which comprised PLEC M6, along with an adjacent vimentin site) interacted with BRCA2. Since each PLEC module exhibits high homology to the others, the possibility of all six domains participating in this interaction was indicated. Moreover, when PLEC M1 was overexpressed in HeLa cells, it competed with endogenous plectin and inhibited the BRCA2-plectin interaction. This inhibitory effect resulted in dissociation of the centrosomes from the nucleus and increased the rate of micronuclei formation which may lead to carcinogenesis. In addition, when either BRCA2 or plectin was suppressed by the appropriate siRNA, a similar change in centrosomal positioning was observed. We suggest that the BRCA2-plectin interaction plays an important role in the regulation of centrosome localization and also that displacement of the centrosome may result in genomic instability and cancer development.


Subject(s)
BRCA2 Protein/physiology , Centrosome/physiology , Plectin/physiology , Apoptosis Regulatory Proteins , CDC2 Protein Kinase/physiology , Cell Line, Tumor , Cell Nucleus/pathology , Cyclin B/physiology , Cyclin B1 , Humans , Immunoprecipitation , Neoplasms/etiology , Plectin/chemistry , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL