Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
Add more filters

Publication year range
1.
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
2.
Mol Cell ; 32(2): 210-20, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18951089

ABSTRACT

Cell-cycle progression is regulated by cyclin-dependent kinases (CDKs). CDK1 and CDK2 can be also activated by noncyclin proteins named RINGO/Speedy, which were identified as inducers of the G2/M transition in Xenopus oocytes. However, it is unclear how XRINGO triggers M phase entry in oocytes. We show here that XRINGO-activated CDKs can phosphorylate specific residues in the regulatory domain of Myt1, a Wee1 family kinase that plays a key role in the G2 arrest of oocytes. We have identified three Ser that are major phosphoacceptor sites for CDK/XRINGO but are poorly phosphorylated by CDK/cyclin. Phosphorylation of these Ser inhibits Myt1 activity, whereas their mutation makes Myt1 resistant to inhibition by CDK/XRINGO. Our results demonstrate that XRINGO-activated CDKs have different substrate specificity than the CDK/cyclin complexes. We also describe a mechanism of Myt1 regulation based on site-specific phosphorylation, which is likely to mediate the induction of G2/M transition in oocytes by XRINGO.


Subject(s)
Cell Cycle Proteins/physiology , Cyclin B/physiology , Cyclin-Dependent Kinases/physiology , DNA-Binding Proteins/metabolism , Meiosis/physiology , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/physiology , Animals , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cell Division/physiology , Cyclin-Dependent Kinase 2/metabolism , DNA-Binding Proteins/chemistry , Down-Regulation , Enzyme Activation , G2 Phase/physiology , Oocytes/cytology , Oocytes/enzymology , Oocytes/metabolism , Phosphorylation , Substrate Specificity , Transcription Factors/chemistry , Xenopus , Xenopus Proteins/chemistry
3.
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
4.
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
5.
Trends Cell Biol ; 16(1): 55-63, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16337124

ABSTRACT

There are two major problems for the cell to solve in mitosis: how to ensure that each daughter cell receives an equal and identical complement of the genome, and how to prevent cell separation before chromosome segregation. Both these problems are solved by controlling when two specific proteins are destroyed: securin, an inhibitor of chromosome segregation, and cyclin B, which inhibits cell separation (cytokinesis). It has recently become clear that several other proteins are degraded at specific points in mitosis. This review (which is part of the Chromosome Segregation and Aneuploidy series) focuses on how specific proteins are selected for proteolysis at defined points in mitosis and how this contributes to the proper coordination of chromosome segregation and cytokinesis.


Subject(s)
Cell Cycle Proteins/physiology , Chromosome Segregation/physiology , Cytokinesis/physiology , Mitosis/physiology , Anaphase-Promoting Complex-Cyclosome , Animals , Cyclin B/physiology , Gene Expression Regulation , Genes, cdc/physiology , Humans , Peptide Hydrolases/physiology , Time Factors , Ubiquitin/physiology , Ubiquitin-Protein Ligase Complexes/physiology
6.
Curr Opin Cell Biol ; 11(2): 267-73, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10209160

ABSTRACT

In eukaryotes, G2/M progression is mediated by activation of mitosis promoting factor (MPF). To ensure faithful chromosome segregation, the activity of key mitotic inducers and inhibitors are coupled with chromosome replication, spindle pole duplication, morphogenesis, and DNA damage. Evidence gathered in the past two years has underscored the importance of positioning MPF and its regulators in the proper place at the proper time to ensure orderly progression through the G2/M transition. Altering the spatial organization of G2/M regulators also contributes to prevention of mitosis following DNA damage.


Subject(s)
CDC2 Protein Kinase/physiology , Cyclin B/physiology , Drosophila Proteins , Maturation-Promoting Factor/physiology , Mitosis/physiology , Animals , Biological Transport , Cell Compartmentation , Cell Cycle/physiology , DNA Damage , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism , Fungal Proteins/physiology , Fungi/cytology , Fungi/physiology , G2 Phase , Humans , Models, Biological , Phosphorylation , Protein Isoforms/physiology , Protein Processing, Post-Translational/physiology , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , Spindle Apparatus/physiology , Vertebrates/physiology , Xenopus laevis/physiology
7.
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
8.
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
9.
Curr Biol ; 17(1): 85-91, 2007 Jan 09.
Article in English | MEDLINE | ID: mdl-17208191

ABSTRACT

Mitosis is thought to be triggered by the activation of Cdk-cyclin complexes. Here we have used RNA interference (RNAi) to assess the roles of three mitotic cyclins, cyclins A2, B1, and B2, in the regulation of centrosome separation and nuclear-envelope breakdown (NEB) in HeLa cells. We found that the timing of NEB was affected very little by knocking down cyclins B1 and B2 alone or in combination. However, knocking down cyclin A2 markedly delayed NEB, and knocking down both cyclins A2 and B1 delayed NEB further. The timing of cyclin B1-Cdk1 activation was normal in cyclin A2 knockdown cells, and there was no delay in centrosome separation, an event apparently controlled by the activation of cytoplasmic cyclin B1-Cdk1. However, nuclear accumulation of cyclin B1-Cdk1 was markedly delayed in cyclin A2 knockdown cells. Finally, a constitutively nuclear cyclin B1, but not wild-type cyclin B1, restored normal NEB timing in cyclin A2 knockdown cells. These findings show that cyclin A2 is required for timely NEB, whereas cyclins B1 and B2 are not. Nevertheless cyclin B1 translocates to the nucleus just prior to NEB in a cyclin A2-dependent fashion and is capable of supporting NEB if rendered constitutively nuclear.


Subject(s)
Centrosome/metabolism , Cyclin A/physiology , Cyclin B/physiology , Mitosis/physiology , Nuclear Envelope/metabolism , Cell Nucleus/metabolism , Cyclin A2 , Cyclin B/metabolism , Cyclin B1 , Cyclin B2 , Cyclin-Dependent Kinases/metabolism , HeLa Cells , Humans
10.
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
11.
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
12.
Carcinogenesis ; 30(9): 1507-16, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19628579

ABSTRACT

BACKGROUND AND AIMS: X-linked inhibitor of apoptosis-associated factor 1 (XAF1) was first recognized as an antagonist of X-linked inhibitor of apoptosis in suppressing caspase 3 activity. It has lower expression in cancer cells than normal tissue. Overexpression of XAF1 can inhibit cancer cell growth and sensitize tumor necrosis factor-related apoptosis-inducing ligand- or etoposide-induced apoptosis. The aim of this study is to elucidate the mechanism of XAF1 in regulating cell growth. METHODS: Stable transfectants of gastrointestinal (GI) cancer cell lines AGS and SW1116 expressing XAF1 and vector control were generated. Cell growth, apoptosis, mitotic status and cell cycle distribution were assessed. The interaction between XAF1 and G(2)/M checkpoint proteins was evaluated by immunoblotting, kinase assay and co-immunoprecipitation assay. Mitotic catastrophe was identified by occurrence of aberrant nuclei and centrosomal amplification. RESULTS: Our results showed that overexpression of XAF1 suppressed serum-dependent cancer cell growth, induced mitotic catastrophe and G(2)/M cell cycle arrest. Interestingly, XAF1 was predominantly expressed in G(2)/M phase after cell cycle synchronization. XAF1 interacted with and activated checkpoint kinase 1 (Chk1), inactivated Cdc25C and lead to inactivation of Cdc2-cyclin B complex. Suppression of Chk1 abrogated XAF1-induced G(2)/M arrest. CONCLUSIONS: Our findings implicate XAF1 as a novel cell cycle modulator that is recruited in G(2)/M phase and thus unravel a novel function pathway of XAF1, suggesting the potential role of XAF1 as the target for the management of GI cancers.


Subject(s)
Gastrointestinal Neoplasms/pathology , Neoplasm Proteins/physiology , Protein Kinases/physiology , Adaptor Proteins, Signal Transducing , Apoptosis , Apoptosis Regulatory Proteins , CDC2 Protein Kinase , Caspase 3/metabolism , Cell Cycle , Cell Division , Cell Line , Cell Proliferation , Checkpoint Kinase 1 , Cyclin B/physiology , Cyclin-Dependent Kinases , DNA Damage , G2 Phase , Humans , Intracellular Signaling Peptides and Proteins , Mitosis
13.
Int J Cancer ; 124(5): 1020-7, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19048596

ABSTRACT

Oncoprotein 18/stathmin (Op18/stathmin) plays a crucial role in maintaining cell biological characteristics by regulating microtubule dynamics, especially entry into mitosis; phosphorylated Op18/stathmin promotes microtubule polymerization to form the mitotic spindle, which is essential for chromosome segregation and cell division. Cdc2 is a critical kinase in starting M phase events in cell-cycle progression and is a positive regulator of the cell cycle. Latent membrane protein 1 (LMP1) is an Epstein-Barr virus (EBV)-encoded oncogenic protein that is able to induce carcinogenesis via various signaling pathways. This study focused on regulation by LMP1 of Op18/stathmin signaling in nasopharyngeal carcinoma (NPC) cells and showed that LMP1 regulates Op18/stathmin signaling through cdc2 mediation, LMP1 upregulates cdc2 kinase activity, and Op18/stathmin phosphorylation promotes the interaction of cdc2 with Op18/stathmin and microtubule polymerization during mitosis, and inhibition of LMP1 expression attenuates the interaction of cdc2 and Op18/stathmin and promotes microtubule depolymerization. These results reveal a new pathway via which LMP1 regulates Op18/stathmin signaling by cdc2 mediation; this new signaling pathway not only perfects the LMP1 regulation network but also elucidates the molecular mechanism of LMP1 that leads to carcinogenesis.


Subject(s)
Cyclin B/physiology , Nasopharyngeal Neoplasms/pathology , Signal Transduction/physiology , Stathmin/physiology , Viral Matrix Proteins/physiology , CDC2 Protein Kinase , Cell Cycle , Cell Line, Tumor , Cyclin B/antagonists & inhibitors , Cyclin-Dependent Kinases , DNA, Catalytic/pharmacology , Herpesvirus 4, Human , Humans , Microtubules/physiology , Phosphorylation
14.
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
15.
J Cell Biol ; 140(2): 283-93, 1998 Jan 26.
Article in English | MEDLINE | ID: mdl-9442104

ABSTRACT

We show that a splice variant-derived cyclin B is produced in sea urchin oocytes and embryos. This splice variant protein lacks highly conserved sequences in the COOH terminus of the protein. It is found strikingly abundant in growing oocytes and cells committed to differentiation during embryogenesis. Cyclin B splice variant (CBsv) protein associates weakly in the cell with Xenopus cdc2 and with budding yeast CDC28p. In contrast to classical cyclin B, CBsv very poorly complements a triple CLN deletion in budding yeast, and its microinjection prevents an initial step in MPF activation, leading to an important delay in oocyte meiosis reinitiation. CBsv microinjection in fertilized eggs induces cell cycle delay and abnormal development. We assume that CBsv is produced in growing oocytes to keep them in prophase, and during embryogenesis to slow down cell cycle in cells that will be committed to differentiation.


Subject(s)
Alternative Splicing , Cyclin B/genetics , Cyclin B/physiology , Sea Urchins/embryology , Amino Acid Sequence , Animals , CDC2 Protein Kinase/metabolism , Cloning, Molecular , Maturation-Promoting Factor/metabolism , Mitosis , Molecular Sequence Data , Oocytes/cytology , RNA, Messenger/chemistry , Sequence Alignment
16.
J Cell Biol ; 146(5): 1019-32, 1999 Sep 06.
Article in English | MEDLINE | ID: mdl-10477756

ABSTRACT

In the budding yeast Saccharomyces cerevisiae, the mitotic spindle must align along the mother-bud axis to accurately partition the sister chromatids into daughter cells. Previous studies showed that spindle orientation required both astral microtubules and the actin cytoskeleton. We now report that maintenance of correct spindle orientation does not depend on F-actin during G2/M phase of the cell cycle. Depolymerization of F-actin using Latrunculin-A did not perturb spindle orientation after this stage. Even an early step in spindle orientation, the migration of the spindle pole body (SPB), became actin-independent if it was delayed until late in the cell cycle. Early in the cell cycle, both SPB migration and spindle orientation were very sensitive to perturbation of F-actin. Selective disruption of actin cables using a conditional tropomyosin double-mutant also led to defects in spindle orientation, even though cortical actin patches were still polarized. This suggests that actin cables are important for either guiding astral microtubules into the bud or anchoring them in the bud. In addition, F-actin was required early in the cell cycle for the development of the actin-independent spindle orientation capability later in the cell cycle. Finally, neither SPB migration nor the switch from actin-dependent to actin-independent spindle behavior required B-type cyclins.


Subject(s)
Actins/metabolism , Cell Cycle , Cell Polarity , Microfilament Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/cytology , Spindle Apparatus/metabolism , Anaphase/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CDC28 Protein Kinase, S cerevisiae/genetics , CDC28 Protein Kinase, S cerevisiae/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Cell Cycle/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Polarity/drug effects , Cyclin B/genetics , Cyclin B/physiology , Dyneins/genetics , Dyneins/physiology , Fungal Proteins/genetics , Fungal Proteins/physiology , Genes, Fungal/genetics , Genes, Fungal/physiology , Hydroxyurea/pharmacology , Microtubules/drug effects , Microtubules/metabolism , Mutation , Nocodazole/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Spindle Apparatus/drug effects , Temperature , Thiazoles/pharmacology , Thiazolidines , Tropomyosin/genetics , Tropomyosin/metabolism
17.
Arch Virol ; 154(7): 1143-9, 2009.
Article in English | MEDLINE | ID: mdl-19526192

ABSTRACT

The conserved murine gammaherpesvirus 68 ORF20 has recently been demonstrated to induce G2 cell cycle arrest followed by apoptosis in human and mouse cells. Here, we demonstrate that its homologues UL24 in HSV-1, ORF20 in KSHV and UL76 in HCMV are also inducers of cell cycle arrest followed by apoptosis in both human and mouse cells. The mechanism of action is similar to that reported for MHV-68 ORF20, inactivating the mitotic complex cyclinB/Cdc2.


Subject(s)
Alphaherpesvirinae/genetics , Betaherpesvirinae/genetics , Gammaherpesvirinae/genetics , Viral Proteins/genetics , 3T3 Cells/cytology , Animals , CDC2 Protein Kinase , Cell Cycle , Cell Line , Conserved Sequence , Cyclin B/physiology , Cyclin-Dependent Kinases , Gene Expression Regulation, Viral , Genes, Reporter , Green Fluorescent Proteins/genetics , Herpesvirus 8, Human/genetics , Humans , Mice , Multigene Family , Open Reading Frames , T-Lymphocytes/cytology
18.
Mol Biol Cell ; 17(2): 814-23, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16339075

ABSTRACT

The mammalian bromodomain protein Brd4 interacts with mitotic chromosomes by binding to acetylated histone H3 and H4 and is thought to play a role in epigenetic memory. Mitotic cells are susceptible to antimicrotubule drugs. These drugs activate multiple response pathways and arrest cells at mitosis. We found that Brd4 was rapidly released from chromosomes upon treatment with antimicrotubule drugs, including the reversible agent nocodazole. Yet, when nocodazole was withdrawn, Brd4 was reloaded onto chromosomes, and cells proceeded to complete cell division. However, cells in which a Brd4 allele was disrupted (Brd4+/-), and expressing only half of the normal Brd4 levels, were defective in reloading Brd4 onto chromosomes. Consequently, Brd4+/- cells were impaired in their ability to recover from nocodazole-induced mitotic arrest: a large fraction of +/- cells failed to reach anaphase after drug withdrawal, and those that entered anaphase showed an increased frequency of abnormal chromosomal segregation. The reloading defect observed in Brd4+/- cells coincided with selective hypoacetylation of lysine residues on H3 and H4. The histone deacetylase inhibitor trichostatin A increased global histone acetylation and perturbed nocodazole-induced Brd4 unloading. Brd4 plays an integral part in a cellular response to drug-induced mitotic stress by preserving a properly acetylated chromatin status.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Microtubules/drug effects , Mitosis/drug effects , Nocodazole/pharmacology , Oncogene Proteins, Fusion/physiology , Acetylation , Animals , Apoptosis/drug effects , CDC2 Protein Kinase/physiology , Cell Proliferation/drug effects , Chromosome Segregation/drug effects , Cyclin B/physiology , Cyclin B1 , Heterozygote , Humans , Hydroxamic Acids/pharmacology , Mice , Mitosis/physiology , Mutagens/pharmacology , Nuclear Proteins , Oncogene Proteins, Fusion/genetics , Polyploidy , Transcription Factors
19.
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
20.
Oncogene ; 26(51): 7175-84, 2007 Nov 08.
Article in English | MEDLINE | ID: mdl-17533373

ABSTRACT

Cyclin-dependent kinase 1 (CDK1) plays a crucial role in establishing metaphase and has also been shown to prevent DNA re-replication. Cyclins B1 and B2 are two known activators of CDK1 operating during mitosis in human cells. Little is known about the specific roles of each of these cyclins in CDK1 activation, but cyclin B2 is thought to play a minor role and to be unable to replace cyclin B1 for mitosis completion. In our study, we found that severe reduction by separate RNA interference of either cyclin B1 or cyclin B2 protein levels results in little or no alteration of the cell cycle and, more specifically, of mitosis progression. In contrast, simultaneous depletion of both B-type cyclins leads to massive accumulation of 4N cells, mitotic failure, premature mitosis exit and DNA re-replication. These defects can be corrected by the ectopic expression of a cyclin B2 resistant to the short hairpin RNA. Altogether, these data show that, in cycling human cells, cyclin B2 can compensate for the downregulation of cyclin B1 during mitosis. They also clearly implicate cyclins B1 and B2 as crucial activators of CDK1 in its biological function of DNA re-replication prevention.


Subject(s)
Cyclin B/physiology , DNA Replication/physiology , Mitosis/physiology , Base Sequence , Blotting, Western , Cell Line , Cyclin B/genetics , Cyclin B1 , Cyclin B2 , DNA Primers , Humans , Polyploidy , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL