Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Proc Natl Acad Sci U S A ; 107(7): 2932-7, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20133761

RESUMO

Centrosomes have recently emerged as key regulators of the cell cycle. The G1/S transition requires a functional centrosome, and centrosomal localization of numerous proteins, including cyclin/Cdk complexes, is important for the G2/M transition. Here we identify a modular centrosomal localization signal (CLS) localizing cyclin A to centrosomes independently of Cdk binding. The cyclin A CLS is located in a distinct part of the molecule compared with the cyclin E CLS and includes the MRAIL hydrophobic patch involved in substrate recognition. The cyclin A CLS interacts with p27(KIP1), and expression of p27(KIP1) removes cyclin A but not cyclin E from centrosomes. Expression of the cyclin A CLS displaces both endogenous cyclin A and E from centrosomes and inhibits DNA replication, supporting an emerging concept that DNA replication is linked to centrosomal events. Structural analysis indicates that differences in surface charge and length of the C-terminal helix explain why the MRAIL region in cyclin E is not a functional CLS. These results indicate that the cyclin A CLS may contribute to targeting and recognition of centrosomal Cdk substrates and is required for specific effects of p27(KIP1) on cyclin A-Cdk2.


Assuntos
Ciclo Celular/fisiologia , Centrossomo/metabolismo , Ciclina A/química , Ciclina A/metabolismo , Modelos Moleculares , Transdução de Sinais/fisiologia , Animais , Western Blotting , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Eletroforese em Gel de Poliacrilamida , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Estrutura Terciária de Proteína , Xenopus
2.
Trends Biochem Sci ; 33(11): 511-3, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18818085

RESUMO

Polo-like kinase 1 (Plk1) is essential for checkpoint recovery and the activation of key mitotic enzymes; however, its own activation mechanism has remained elusive. Recent findings show that Bora, a G(2)-M expressed protein, facilitates Plk1 activation by the oncogenic kinase Aurora A in G(2). During mitosis, Plk1-dependent Bora degradation promotes Aurora A localization to the centrosome and/or spindle. Bora-dependent regulation provides important new insights into interactions between key mitotic kinases.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Aurora Quinases , Proteínas de Drosophila/fisiologia , Ativação Enzimática , Retroalimentação Fisiológica , Modelos Biológicos , Quinase 1 Polo-Like
3.
J Cell Sci ; 123(Pt 16): 2743-9, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20663915

RESUMO

Centrosomes are the major microtubule-organizing centers in animal cells and regulate formation of a bipolar mitotic spindle. Aberrant centrosome number causes chromosome mis-segregation, and has been implicated in genomic instability and tumor development. Previous studies have demonstrated a role for the DNA replication factors MCM5 and Orc1 in preventing centrosome reduplication. Cyclin A-Cdk2 localizes on centrosomes by means of a modular centrosomal localization sequence (CLS) that is distinct from that of cyclin E. Here, we show that cyclin A interacts with both MCM5 and Orc1 in a CLS-dependent but Cdk-independent manner. Although the MRAIL hydrophobic patch is contained within the cyclin A CLS, binding of both MCM5 and Orc1 to cyclin A does not require a wild-type hydrophobic patch. The same domain in MCM5 that mediates interaction with cyclin E also binds cyclin A, resulting in centrosomal localization of MCM5. Finally, unlike its function in DNA synthesis, MCM5-mediated inhibition of centrosome reduplication in S-phase-arrested CHO cells does not require binding to other MCM family members. These results suggest that cyclins E and A sequentially prevent centrosome reduplication throughout interphase by recruitment of DNA replication factors such as MCM5 and Orc1.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Centrossomo/fisiologia , Ciclina A/fisiologia , Complexo de Reconhecimento de Origem/fisiologia , Animais , Células CHO , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Cricetinae , Cricetulus , Ciclina A/genética , Ciclina A/metabolismo , Imunofluorescência , Células HeLa , Humanos , Imunoprecipitação , Centro Organizador dos Microtúbulos/metabolismo , Complexo de Reconhecimento de Origem/genética , Complexo de Reconhecimento de Origem/metabolismo , Transfecção
4.
Curr Biol ; 18(7): 519-25, 2008 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-18372177

RESUMO

Targeting protein for Xklp2 (TPX2) activates the Ser/Thr kinase Aurora A in mitosis and targets it to the mitotic spindle [1, 2]. These effects on Aurora A are mediated by the N-terminal domain of TPX2, whereas a C-terminal fragment has been reported to affect microtubule nucleation [3]. Using the Xenopus system, we identified a novel role of TPX2 during mitosis. Injection of TPX2 or its C terminus (TPX2-CT) into blastomeres of two-cell embryos led to potent cleavage arrest. Despite cleavage arrest, TPX2-injected embryos biochemically undergo multiple rounds of DNA synthesis and mitosis, and arrested blastomeres have abnormal spindles, clustered centrosomes, and an apparent failure of cytokinesis. In Xenopus S3 cells, transfection of TPX2-FL causes spindle collapse, whereas TPX2-CT blocks pole segregation, resulting in apposing spindle poles with no evident displacement of Aurora A. Analysis of TPX2-CT deletion peptides revealed that only constructs able to interact with the class 5 kinesin-like motor protein Eg5 induce the spindle phenotypes. Importantly, injection of Eg5 into TPX2-CT-arrested blastomeres causes resumption of cleavage. These results define a discrete domain within the C terminus of TPX2 that exerts a novel Eg5-dependent function in spindle pole segregation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Desenvolvimento Embrionário/fisiologia , Cinesinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/fisiologia , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fuso Acromático/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Linhagem Celular , Embrião não Mamífero/metabolismo , Embrião não Mamífero/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , Fuso Acromático/fisiologia , Xenopus
5.
Mol Cell Biol ; 27(19): 6852-62, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17664286

RESUMO

In Xenopus laevis embryos, the midblastula transition (MBT) at the 12th cell division marks initiation of critical developmental events, including zygotic transcription and the abrupt inclusion of gap phases into the cell cycle. Interestingly, although an ionizing radiation-induced checkpoint response is absent in pre-MBT embryos, introduction of a threshold amount of undamaged plasmid or sperm DNA allows a DNA damage checkpoint response to be activated. We show here that undamaged threshold DNA directly participates in checkpoint signaling, as judged by several dynamic changes, including H2AX phosphorylation, ATM phosphorylation and loading onto chromatin, and Chk1/Chk2 phosphorylation and release from nuclear DNA. These responses on physically separate threshold DNA require gamma-H2AX and are triggered by an ATM-dependent soluble signal initiated by damaged DNA. The signal persists in egg extracts even after damaged DNA is removed from the system, indicating that the absence of damaged DNA is not sufficient to end the checkpoint response. The results identify a novel mechanism by which undamaged DNA enhances checkpoint signaling and provide an example of how the transition to cell cycle checkpoint activation during development is accomplished by maternally programmed increases in the DNA-to-cytoplasm ratio.


Assuntos
Ciclo Celular/fisiologia , Dano ao DNA , DNA/metabolismo , Embrião não Mamífero/fisiologia , Genes cdc , Transdução de Sinais/fisiologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Embrião não Mamífero/anatomia & histologia , Histonas/genética , Histonas/metabolismo , Masculino , Oócitos/fisiologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Espermatozoides/fisiologia , Extratos de Tecidos/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/genética
6.
Curr Biol ; 16(19): 1968-73, 2006 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-17027495

RESUMO

Cytostatic factor (CSF) arrests vertebrate eggs in metaphase of meiosis II through several pathways that inhibit activation of the anaphase-promoting complex/cyclosome (APC/C). In Xenopus, the Mos-MEK1-MAPK-p90(Rsk) cascade utilizes spindle-assembly-checkpoint components to effect metaphase arrest. Another pathway involves cyclin E-Cdk2, and sustained cyclin E-Cdk2 activity in egg extracts causes metaphase arrest in the absence of Mos; this latter finding suggests that an independent pathway contributes to CSF arrest. Here, we demonstrate that metaphase arrest with cyclin E-Cdk2, but not with Mos, requires the spindle-checkpoint kinase monopolar spindles 1 (Mps1), a cyclin E-Cdk2 target that is also implicated in centrosome duplication. xMps1 is synthesized and activated during oocyte maturation and inactivated upon CSF release. In egg extracts, CSF release by calcium was inhibited by constitutively active cyclin E-Cdk2 and delayed by wild-type xMps1. Ablation of cyclin E by antisense oligonucleotides blocked accumulation of xMps1, suggesting that cyclin E-Cdk2 controls Mps1 levels. During meiosis II, activated cyclin E-Cdk2 significantly inhibited the APC/C even in the absence of the Mos-MAPK pathway, but this inhibition was not sufficient to suppress S phase between meiosis I and II. These results uniquely place xMps1 downstream of cyclin E-Cdk2 in mediating a pathway of APC/C inhibition and metaphase arrest.


Assuntos
Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Metáfase/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas de Xenopus/fisiologia , Animais , Ciclina E/antagonistas & inibidores , Ciclina E/genética , Sistema de Sinalização das MAP Quinases , Oligonucleotídeos Antissenso , Oócitos/citologia , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-mos/metabolismo , Proteínas Proto-Oncogênicas c-mos/fisiologia , Xenopus , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
7.
Dev Cell ; 7(2): 275-81, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15296723

RESUMO

In Xenopus, cell cycle checkpoints monitoring DNA damage, DNA replication, and spindle assembly do not appear until after the midblastula transition (MBT; 4000 cells). We show that a DNA damage checkpoint can slow the cell cycle even in 2-cell embryos when the DNA content is increased. Slowing follows caffeine-sensitive activation of the checkpoint kinase, Chk1; degradation of the cell cycle phosphatase, Cdc25A; and inhibitory phosphorylation of Cdc25C and cyclin-dependent kinases (Cdks). Alterations in the DNA-to-cytoplasmic ratio elicit a dose-dependent DNA damage checkpoint, and the ratio required to activate Chk1 for the damage response is lower than that associated with "developmental" activation of Chk1 shortly after the MBT. Our results indicate that a maternal damage response, independent of zygotic transcription, is present even in very early embryos, and requires both double-stranded DNA ends and a threshold DNA-to-cytoplasmic ratio to significantly affect the cell cycle.


Assuntos
Citoplasma/metabolismo , Dano ao DNA , Animais , Cafeína/farmacologia , Ciclo Celular , Estimulantes do Sistema Nervoso Central/farmacologia , Quinase 1 do Ponto de Checagem , DNA/química , DNA/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Immunoblotting , Fosforilação , Proteínas Quinases/metabolismo , Fatores de Tempo , Transcrição Gênica , Xenopus , Proteínas de Xenopus , Fosfatases cdc25/metabolismo
8.
J Cell Biol ; 163(6): 1231-42, 2003 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-14691134

RESUMO

In cells containing disrupted spindles, the spindle assembly checkpoint arrests the cell cycle in metaphase. The budding uninhibited by benzimidazole (Bub) 1, mitotic arrest-deficient (Mad) 1, and Mad2 proteins promote this checkpoint through sustained inhibition of the anaphase-promoting complex/cyclosome. Vertebrate oocytes undergoing meiotic maturation arrest in metaphase of meiosis II due to a cytoplasmic activity termed cytostatic factor (CSF), which appears not to be regulated by spindle dynamics. Here, we show that microinjection of Mad1 or Mad2 protein into early Xenopus laevis embryos causes metaphase arrest like that caused by Mos. Microinjection of antibodies to either Mad1 or Mad2 into maturing oocytes blocks the establishment of CSF arrest in meiosis II, and immunodepletion of either protein blocked the establishment of CSF arrest by Mos in egg extracts. A Mad2 mutant unable to oligomerize (Mad2 R133A) did not cause cell cycle arrest in blastomeres or in egg extracts. Once CSF arrest has been established, maintenance of metaphase arrest requires Mad1, but not Mad2 or Bub1. These results suggest a model in which CSF arrest by Mos is mediated by the Mad1 and Mad2 proteins in a manner distinct from the spindle checkpoint.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Meiose/fisiologia , Metáfase/fisiologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-mos/metabolismo , Proteínas Repressoras/metabolismo , Animais , Anticorpos/farmacologia , Proteínas de Ligação ao Cálcio/farmacologia , Proteínas de Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Feminino , Genes cdc/efeitos dos fármacos , Genes cdc/fisiologia , Proteínas Mad2 , Meiose/efeitos dos fármacos , Metáfase/efeitos dos fármacos , Mutação/genética , Proteínas Nucleares , Proteínas Oncogênicas v-mos/genética , Proteínas Oncogênicas v-mos/metabolismo , Oócitos/citologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oogênese/efeitos dos fármacos , Oogênese/fisiologia , Fosfoproteínas/farmacologia , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Repressoras/farmacologia , Xenopus laevis
10.
Curr Biol ; 15(16): 1458-68, 2005 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-16040245

RESUMO

BACKGROUND: Vertebrate oocytes are arrested at second meiotic metaphase by cytostatic factor (CSF) while awaiting fertilization. Accumulating evidence has suggested that inhibition of the anaphase-promoting complex/cyclosome (APC/C) is responsible for this arrest. Xenopus polo-like kinase 1 (Plx1) is required for activation of the APC/C at the metaphase-anaphase transition, and calcium elevation, upon fertilization/activation of eggs, acting through calmodulin-dependent kinase II (CaMKII) is sufficient to activate the APC/C and terminate CSF arrest. However, connections between the Plx1 pathway and the CaMKII pathway have not been identified. RESULTS: Overexpression of Plx1 causes CSF release in the absence of calcium, and depletion of Plx1 from egg extracts blocks induction of CSF release by calcium and CaMKII. Prior phosphorylation of the APC/C inhibitor XErp1/Emi2 by CaMK II renders it a good substrate for Plx1, and phosphorylation by both kinases together promotes its degradation in egg extracts. The pathway is enhanced by the ability of Plx1 to cause calcium-independent activation of CaMKII. The results identify the targets of CaMKII and Plx1 that promote egg activation and define the first known pathway of CSF release in which an APC/C inhibitor is targeted for degradation only when both CaMKII and Plx1 are active after calcium elevation at fertilization. CONCLUSIONS: Plx1 with an intact polo-box domain is necessary for release of CSF arrest and sufficient when overexpressed. It acts at the same level as CaMKII in the pathway of calcium-induced CSF release by cooperating with CaMKII to regulate APC/C regulator(s), such as XErp1/Emi2, rather than by directly activating the APC/C itself.


Assuntos
Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Fertilização/fisiologia , Metáfase/fisiologia , Oócitos/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Xenopus/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas de Ciclo Celular/genética , Núcleo Celular/ultraestrutura , Clonagem Molecular , Primers do DNA , Immunoblotting , Masculino , Oócitos/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-mos/metabolismo , Espermatozoides/ultraestrutura , Complexos Ubiquitina-Proteína Ligase/metabolismo , Xenopus , Proteínas de Xenopus/genética
11.
Mol Endocrinol ; 21(3): 664-73, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17185392

RESUMO

Rapid, nongenomic membranal effects of progesterone were demonstrated in amphibian oocytes more than 30 y ago. Recently, a distinct family of membrane progestin receptors (mPRs) has been cloned in fish and other vertebrate species. In this study we explore the role of mPR in promoting oocyte maturation in Xenopus laevis. RT-PCR analysis indicates that Xenopus oocytes contain transcripts for the mPRbeta ortholog, similar to what has been reported in zebrafish oocytes, and Western blotting shows that the protein is expressed on the oocyte plasma membrane. Microinjection of mPRbeta-specific antibodies into oocytes resulted in a dramatic inhibition of progesterone-dependent oocyte maturation, whereas microinjection of mRNA encoding Myc-Xenopus mPR (XmPR)beta resulted in an accelerated rate of progesterone-induced oocyte maturation, concomitant with membranal localization of the protein. Binding studies in mammalian cells expressing XmPRbeta confirmed specific binding of progesterone by the expressed protein. These results suggest that XmPRbeta is a physiological progesterone receptor involved in initiating the resumption of meiosis during maturation of Xenopus oocytes.


Assuntos
Oócitos/efeitos dos fármacos , Progesterona/farmacologia , Receptores de Progesterona/fisiologia , Xenopus laevis , Animais , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Proteínas de Membrana/fisiologia , Oócitos/metabolismo , Ligação Proteica , Receptores de Progesterona/metabolismo , Maturidade Sexual/efeitos dos fármacos , Transfecção
12.
Curr Biol ; 12(12): 1027-33, 2002 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-12123578

RESUMO

In vertebrate unfertilized eggs, metaphase arrest in Meiosis II is mediated by an activity known as cytostatic factor (CSF). CSF arrest is dependent upon Mos-dependent activation of the MAPK/Rsk pathway, and Rsk activates the spindle checkpoint kinase Bub1, leading to inhibition of the anaphase-promoting complex (APC), an E3 ubiquitin ligase required for the metaphase/anaphase transition. However, it is not known whether Bub1 is required for the establishment of CSF arrest or whether other pathways also contribute. Here, we show that immunodepletion of Bub1 from egg extracts blocks the ability of Mos to establish CSF arrest, and arrest can be restored by the addition of wild-type, but not kinase-dead, Bub1. The appearance of CSF arrest at Meiosis II may result from coexpression of cyclin E/Cdk2 with the MAPK/Bub1 pathway. Cyclin E/Cdk2 was able to cause metaphase arrest in egg extracts even in the absence of Mos and could also inhibit cyclin B degradation in oocytes when expressed at anaphase of Meiosis I. Once it has been established, metaphase arrest can be maintained in the absence of MAPK, Bub1, or cyclin E/Cdk2 activity. Both pathways are independent of each other, but each appears to block activation of the APC, which is required for cyclin B degradation and the metaphase/anaphase transition.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Ciclina E/fisiologia , Quinases Ciclina-Dependentes/fisiologia , Meiose/fisiologia , Proteínas Quinases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-mos/fisiologia , Animais , Linhagem Celular , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/genética , Mutação , Proteínas Serina-Treonina Quinases/genética , Xenopus , Proteínas de Xenopus
13.
Curr Biol ; 13(8): 691-7, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12699628

RESUMO

Segregation of chromosomes during mitosis requires interplay between several classes of protein on the spindle, including protein kinases, protein phosphatases, and microtubule binding motor proteins [1-4]. Aurora A is an oncogenic cell cycle-regulated protein kinase that is subject to phosphorylation-dependent activation [5-11]. Aurora A localization to the mitotic spindle depends on the motor binding protein TPX2 (Targeting Protein for Xenopus kinesin-like protein 2), but the protein(s) involved in Aurora A activation are unknown [11-13]. Here, we purify an activator of Aurora A from Xenopus eggs and identify it as TPX2. Remarkably, Aurora A that has been fully deactivated by Protein Phosphatase 2A (PP2A) becomes phosphorylated and reactivated by recombinant TPX2 in an ATP-dependent manner. Increased phosphorylation and activation of Aurora A requires its own kinase activity, suggesting that TPX2 stimulates autophosphorylation and autoactivation of the enzyme. Consistently, wild-type Aurora A, but not a kinase inactive mutant, becomes autophosphorylated on the regulatory T loop residue (Thr 295) after TPX2 treatment. Active Aurora A from bacteria is further activated at least 7-fold by recombinant TPX2, and TPX2 also impairs the ability of protein phosphatases to inactivate Aurora A in vitro. This concerted mechanism of stimulation of activation and inhibition of deactivation implies that TPX2 is the likely regulator of Aurora A activity at the mitotic spindle and may explain why loss of TPX2 in model systems perturbs spindle assembly [14-16]. Our finding that a known binding protein, and not a conventional protein kinase, is the relevant activator for Aurora A suggests a biochemical model in which the dynamic localization of TPX2 on mitotic structures directly modulates the activity of Aurora A for spindle assembly.


Assuntos
Proteínas de Ciclo Celular , Reativadores Enzimáticos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/fisiologia , Proteínas de Neoplasias , Proteínas Nucleares , Fosfoproteínas , Proteínas Quinases/metabolismo , Fuso Acromático/fisiologia , Proteínas de Xenopus , Animais , Aurora Quinases , Autorradiografia , Segregação de Cromossomos/fisiologia , Proteínas Associadas aos Microtúbulos/isolamento & purificação , Fosforilação , Proteínas Serina-Treonina Quinases , Xenopus
14.
Mol Cell Biol ; 24(24): 10573-83, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15572664

RESUMO

We investigated the role of Rsk proteins in the nerve growth factor (NGF) signaling pathway in PC12 cells. When rat Rsk1 or murine Rsk2 proteins were transiently expressed, NGF treatment (100 ng/ml for 3 days) caused three- and fivefold increases in Rsk1 and Rsk2 activities, respectively. Increased activation of both wild-type Rsk proteins could be achieved by coexpression of a constitutively active (CA) mitogen-activated protein kinase (MAPK) kinase, MEK1-DD, which is known to cause differentiation of PC12 cells even in the absence of NGF. Rsk1 and Rsk2 mutated in the PDK1-binding site were not activated by either NGF or MEK1-DD. Expression of constitutively active Rsk1 or Rsk2 in PC12 cells resulted in highly active proteins whose levels of activity did not change either with NGF treatment or after coexpression with MEK1-DD. Rsk2-CA expression had no detectable effect on the cells. However, expression of Rsk1-CA led to differentiation of PC12 cells even in the absence of NGF, as evidenced by neurite outgrowth. Differentiation was not observed with a nonactive Rsk1-CA that was mutated in the PDK1-binding site. Expression of Rsk1-CA did not lead to activation of the endogenous MAPK pathway, indicating that Rsk1 is sufficient to induce neurite outgrowth and is the only target of MAPK required for this effect. Collectively, our data demonstrate a key role for Rsk1 in the differentiation process of PC12 cells.


Assuntos
Diferenciação Celular , Neurônios/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Embrião não Mamífero , Ativação Enzimática , Feminino , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , MAP Quinase Quinase 1/metabolismo , Microinjeções , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Oócitos/metabolismo , Técnicas de Cultura de Órgãos , Células PC12 , RNA Mensageiro/metabolismo , Ratos , Proteínas Quinases S6 Ribossômicas 90-kDa/análise , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Transdução de Sinais , Fatores de Tempo , Xenopus
15.
Oncogene ; 24(2): 238-47, 2005 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-15640839

RESUMO

The Xenopus Polo-like kinase Plx1 plays multiple roles in mitosis. Accumulating evidence shows that Plx1 is the trigger kinase for the G2/M transition that phosphorylates and activates the phosphatase Cdc25C, which subsequently dephosphorylates Cdc2/cyclin B and initiates a positive feedback loop between Cdc25C and Cdc2/cyclin B. Recent findings indicate that Plx1 itself is also in a positive feedback loop. It phosphorylates and activates the protein kinase xPlkk1, which itself then phosphorylates and further activates Plx1. Plx1 functions on the centrosome to promote bipolar spindle formation. Plx1 associates with the anaphase-promoting complex/cyclosome (APC/C) and is required to activate the APC/C for degradation of mitotic regulators required for sister chromatid separation and exit from mitosis. Plx1 is also required for cytokinesis and is localized on the midbody of the contractile ring. All known functions of Plx1 require not only its kinase activity but also an intact polo box domain in the C-terminus.


Assuntos
Mitose/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas de Xenopus/fisiologia , Animais , Proteínas de Ciclo Celular , Centrossomo/fisiologia , Cromátides/fisiologia , Citocinese/fisiologia , Retroalimentação Fisiológica/fisiologia , Fase G2/fisiologia , Fuso Acromático/fisiologia , Xenopus
16.
Mol Cell Endocrinol ; 187(1-2): 173-8, 2002 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-11988325

RESUMO

A cytoplasmic activity in mature oocytes responsible for second meiotic metaphase arrest was identified over 30 years ago in amphibian oocytes. In Xenopus oocytes cytostatic factor (CSF) activity is initiated by the progesterone-dependent synthesis of Mos, a MAPK kinase kinase that activates the MAPK pathway. CSF arrest is mediated by a sole MAPK target, the protein kinase p90(Rsk). Rsk phosphorylates and activates the Bub1 protein kinase, which may cause metaphase arrest due to inhibition of the anaphase-promoting complex (APC) by a conserved mechanism defined genetically in yeast and mammalian cells. CSF arrest in vertebrate oocytes by p90(Rsk) provides a link between the MAPK pathway and the spindle assembly checkpoint in the cell cycle.


Assuntos
Meiose , Oócitos/citologia , Proteínas Proto-Oncogênicas c-mos/fisiologia , Vertebrados/fisiologia , Animais , Ciclo Celular/fisiologia , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Oócitos/metabolismo
17.
Cell Cycle ; 10(2): 199-205, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21217199

RESUMO

Recent identification of the modular CLS motifs responsible for cyclins A and E localization on centrosomes has revealed a tight linkage between the nuclear and centrosomal cycles. These G1/S cyclins must localize on the centrosome in order for DNA replication to occur in the nucleus, whereas essential DNA replication factors also function on the centrosome to prevent centrosome overduplication. Both events are dependent on the presence of an intact CLS within each cyclin. Here we compare the cyclins A and E CLSs at the structural and functional levels and identify a new cyclin A CLS mutant that disrupts all CLS functions and reduces the affinity of cyclin A for Cdk2. Analysis of interactions of the CLS motif within the cyclin molecules highlights the importance of the cyclin CBOX1 region for Cdk2 binding.


Assuntos
Centrossomo/metabolismo , Ciclina A/análise , Ciclina E/análise , Sequência de Aminoácidos , Animais , Células CHO , Centrossomo/química , Cricetinae , Cricetulus , Ciclina A/metabolismo , Ciclina A/fisiologia , Ciclina E/metabolismo , Ciclina E/fisiologia , Quinase 2 Dependente de Ciclina/metabolismo , Fase G1 , Dados de Sequência Molecular , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Fase S
18.
Curr Biol ; 21(5): 428-32, 2011 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-21353560

RESUMO

Supernumerary centrosomes are a key cause of genomic instability in cancer cells. New centrioles can be generated by duplication with a mother centriole as a platform or, in the absence of preexisting centrioles, by formation de novo. Polo-like kinase 4 (Plk4) regulates both modes of centriole biogenesis, and Plk4 deregulation has been linked to tumor development. We show that Plx4, the Xenopus homolog of mammalian Plk4 and Drosophila Sak, induces de novo centriole formation in vivo in activated oocytes and in egg extracts, but not in immature or in vitro matured oocytes. Both kinase activity and the polo-box domain of Plx4 are required for de novo centriole biogenesis. Polarization microscopy in "cycling" egg extracts demonstrates that de novo centriole formation is independent of Cdk2 activity, a major difference compared to template-driven centrosome duplication that is linked to the nuclear cycle and requires cyclinA/E/Cdk2. Moreover, we show that the Mos-MAPK pathway blocks Plx4-dependent de novo centriole formation before fertilization, thereby ensuring paternal inheritance of the centrosome. The results define a new system for studying the biochemical and molecular basis of de novo centriole formation and centriole biogenesis in general.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centríolos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética , Proteínas de Xenopus/metabolismo , Animais , Western Blotting , Microscopia de Polarização , Oócitos/metabolismo , Xenopus
19.
Mol Biol Cell ; 22(13): 2157-64, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21551066

RESUMO

Greatwall kinase has been identified as a key element in M phase initiation and maintenance in Drosophila, Xenopus oocytes/eggs, and mammalian cells. In M phase, Greatwall phosphorylates endosulfine and related proteins that bind to and inhibit protein phosphatase 2A/B55, the principal phosphatase for Cdk-phosphorylated substrates. We show that Greatwall binds active PP2A/B55 in G2 phase oocytes but dissociates from it when progesterone-treated oocytes reach M phase. This dissociation does not require Greatwall kinase activity or phosphorylation at T748 in the presumptive T loop of the kinase. A mutant K71M Greatwall, also known as Scant in Drosophila, induces M phase in the absence of progesterone when expressed in oocytes, despite its reduced stability and elevated degradation by the proteasome. M phase induction by Scant Greatwall requires protein synthesis but is not associated with altered binding or release of PP2A/B55 as compared to wild-type Greatwall. However, in vitro studies with Greatwall proteins purified from interphase cells indicate that Scant, but not wild-type Greatwall, has low but detectable activity against endosulfine. These results demonstrate progesterone-dependent regulation of the PP2A/B55-Greatwall interaction during oocyte maturation and suggest that the cognate Scant Greatwall mutation has sufficient constitutive kinase activity to promote M phase in Xenopus oocytes.


Assuntos
Oócitos/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas de Xenopus/fisiologia , Xenopus/fisiologia , Animais , Divisão Celular , Quinases Ciclina-Dependentes/metabolismo , Feminino , Fase G2 , Peptídeos e Proteínas de Sinalização Intercelular , Mutação , Oócitos/metabolismo , Peptídeos/metabolismo , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Progesterona/metabolismo , Ligação Proteica , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Xenopus/genética , Xenopus/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA