Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
Cell Cycle ; 9(21): 4364-9, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20980823

RESUMO

Activation of the DNA damage response (DDR) is critical for genomic integrity and tumor suppression. The occurrence of DNA damage quickly evokes the DDR through ATM/ATR-dependent signal transduction, which promotes DNA repair and activates the checkpoint to halt cell cycle progression. The "turn off" process of the DDR upon satisfaction of DNA repair, also known as "checkpoint recovery", involves deactivation of DDR elements, but the mechanism is poorly understood. Greatwall kinase (Gwl) has been identified as a key element in the G(2)/M transition and helps maintain M phase through inhibition of PP 2A/B55δ, the principal phosphatase for Cdk-phosphorylated substrates. Here we show that Gwl also promotes recovery from DNA damage and is itself directly inhibited by the DNA damage response (DDR). In Xenopus egg extracts, immunodepletion of Gwl increased the DDR to damaged DNA, whereas addition of wild type, but not kinase dead Gwl, inhibited the DDR. The removal of damaged DNA from egg extracts leads to recovery from checkpoint arrest and entry into mitosis, a process impaired by Gwl depletion and enhanced by Gwl overexpression. Moreover, activation of Cdk1 after the removal of damaged DNA is regulated by Gwl. Collectively, these results defines Gwl as a new regulator of the DDR, which plays an important role in recovery from DNA damage.


Assuntos
Dano ao DNA , Reparo do DNA , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Proteína Quinase CDC2/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Divisão Celular , Fase G2 , Mitose , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiologia , Xenopus laevis
5.
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
6.
Curr Biol ; 20(5): 387-96, 2010 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-20188555

RESUMO

BACKGROUND: In response to DNA damage, cells activate checkpoints to halt cell-cycle progression and prevent genomic instability. Checkpoint activation induced by DNA double-strand breaks (DSB) is dependent on the ATM kinase, a master regulator of the DNA damage response (DDR) that is activated through autophosphorylation and monomerization. RESULTS: Here we show that either protein phosphatase 1 or 2A is sufficient to suppress activation of the DDR and that simultaneous inhibition of both phosphatases fully activates the response. PP1-dependent DDR regulation is mediated by its chromatin-targeting subunit, Repo-Man. Studies in Xenopus egg extracts demonstrate that Repo-Man interacts with ATM and PP1 through distinct domains, leading to PP1-dependent regulation of ATM phosphorylation and activation. Consequently, the level of Repo-Man determines the activation threshold of the DNA damage checkpoint. Repo-Man interacts and extensively colocalizes with ATM in human cells. Expression of wild-type, but not PP1 binding-deficient, Repo-Man attenuates DNA damage-induced ATM activation. Moreover, Repo-Man dissociates from active ATM at DNA damage sites, suggesting that activation of the DDR involves removal of inhibitory regulators. Analysis of primary tumor tissues and cell lines demonstrates that Repo-Man is frequently upregulated in many types of cancers. Elevated Repo-Man expression blunts DDR activation in precancerous cells, whereas knockdown of Repo-Man in malignant cancer cells resensitizes the DDR and restrains growth in soft agar. CONCLUSIONS: We report essential DDR regulation mediated by Repo-Man-PP1 and further delineate underlying mechanisms. Moreover, our evidence suggests that elevated Repo-Man contributes to cancer progression.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Dano ao DNA/fisiologia , Proteínas Nucleares/metabolismo , Proteína Fosfatase 1/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Cromatina , Regulação da Expressão Gênica/fisiologia , Humanos , Proteínas Nucleares/genética , Proteína Fosfatase 2/metabolismo , Xenopus , Proteínas de Xenopus/metabolismo
7.
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
8.
Cell Cycle ; 8(15): 2413-9, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19556869

RESUMO

Entry into mitosis requires the activation of mitotic kinases, including Aurora A and Polo-like kinase 1 (Plk1). Increased levels of these kinases are frequently found associated with human cancers, and therefore it is imperative to understand the processes leading to their activation. We demonstrate that TPX2, but neither Ajuba nor Inhibitor-2, can activate Aurora A directly. Moreover, Plx1 can induce Aurora A T-loop phosphorylation indirectly in vivo during oocyte maturation. We identify Ser204 in TPX2 as a Plx1 phosphorylation site. Mutating Ser204 to alanine decreases activation of Aurora A, whereas a phosphomimetic Asp mutant exhibits enhanced activating ability. Finally, we show that phosphorylation of TPX2 with Plx1 increases its ability to activate Aurora A. Taken together, our data indicate that Plx1 promotes activation of Aurora A, most likely through TPX2. In light of the current literature, we propose a model in which Plx1 and Aurora A activate each other in a positive feedback loop.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Oócitos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animais , Aurora Quinases , Células CHO/metabolismo , Ciclo Celular/fisiologia , Cricetinae , Cricetulus , Fosforilação/fisiologia
9.
J Biol Chem ; 284(9): 5497-505, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19121998

RESUMO

p53 is an important tumor suppressor regulating the cell cycle at multiple stages in higher vertebrates. The p53 gene is frequently deleted or mutated in human cancers, resulting in loss of p53 activity. This leads to centrosome amplification, aneuploidy, and tumorigenesis, three phenotypes also observed after overexpression of the oncogenic kinase Aurora A. Accordingly, recent studies have focused on the relationship between these two proteins. p53 and Aurora A have been reported to interact in mammalian cells, but the function of this interaction remains unclear. We recently reported that Xenopus p53 can inhibit Aurora A activity in vitro but only in the absence of TPX2. Here we investigate the interplay between Xenopus Aurora A, TPX2, and p53 and show that newly synthesized TPX2 is required for nearly all Aurora A activation and for full p53 synthesis and phosphorylation in vivo during oocyte maturation. In vitro, phosphorylation mediated by Aurora A targets serines 129 and 190 within the DNA binding domain of p53. Glutathione S-transferase pull-down studies indicate that the interaction occurs via the p53 transactivation domain and the Aurora A catalytic domain around the T-loop. Our studies suggest that targeting of TPX2 might be an effective strategy for specifically inhibiting the phosphorylation of Aurora A substrates, including p53.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Oócitos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animais , Aurora Quinases , Feminino , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Imunoprecipitação , Oócitos/citologia , Fosforilação , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/genética , Xenopus laevis/crescimento & desenvolvimento
10.
J Cell Sci ; 121(Pt 19): 3224-32, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18799789

RESUMO

Centrosomes are the primary microtubule-organizing centers in animal cells and are required for bipolar spindle assembly during mitosis. Amplification of centrosome number is commonly observed in human cancer cells and might contribute to genomic instability. Cyclin E-Cdk2 has been implicated in regulating centrosome duplication both in Xenopus embryos and extracts and in mammalian cells. Localization of cyclin E on centrosomes is mediated by a 20-amino acid domain termed the centrosomal localization sequence (CLS). In this paper, cyclin E is shown to directly interact with and colocalize on centrosomes with the DNA replication factor MCM5 in a CLS-dependent but Cdk2-independent manner. The domain in MCM5 that is responsible for interaction with cyclin E is distinct from any previously described for MCM5 function and is highly conserved in MCM5 proteins from yeast to mammals. Expression of MCM5 or its cyclin E-interacting domain, but not MCM2, significantly inhibits over-duplication of centrosomes in CHO cells arrested in S-phase. These results indicate that proteins involved in DNA replication might also regulate centrosome duplication.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Ciclina E/metabolismo , Proteínas Oncogênicas/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Proteínas de Ciclo Celular/química , Cricetinae , Cricetulus , Células HeLa , Humanos , Componente 2 do Complexo de Manutenção de Minicromossomo , Modelos Biológicos , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Ligação Proteica , Sinais Direcionadores de Proteínas , Estrutura Terciária de Proteína , Transporte Proteico
11.
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
12.
Mol Cell Biol ; 28(12): 4196-203, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18378691

RESUMO

The dynamics of the Aurora B protein kinase during Xenopus oocyte meiotic maturation were examined. Resting G2 oocytes express inactive Aurora B that is not associated with other subunits of the chromosome passenger complex (CPC). Activity increases near the time of germinal vesicle breakdown in progesterone-treated oocytes, and this increase is correlated with the synthesis of inner centromere protein (INCENP) and survivin, components of the CPC. Ablation of INCENP synthesis led to the failure of progesterone treatment to activate Aurora B, but biochemical progression through the meiosis I-to-II transition and arrest at metaphase II were not affected. At fertilization, Aurora B was deactivated in concert with the degradation of INCENP, and the levels of Aurora B kinase activity and INCENP oscillated in subsequent embryonic cell cycles. Prevention of the decrease in Aurora B activity at fertilization by expression of ectopic wild-type INCENP, but not kinase-dead Aurora B INCENP, blocked calcium-induced exit from metaphase arrest in egg extracts.


Assuntos
Regulação da Expressão Gênica , Oócitos/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Animais , Apoptose , Aurora Quinases , Centrômero/ultraestrutura , Clonagem Molecular , Feminino , Fertilização , Masculino , Modelos Biológicos , Progesterona/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis
13.
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
14.
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
15.
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
16.
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
17.
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
18.
Cell Cycle ; 4(6): 784-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15908779

RESUMO

The Aurora A and B protein kinases are key players in mitotic control and the etiology of human cancer. Despite the near identity of amino acid sequence in the catalytic domain, monomeric Aurora B is 50 fold lower in activity than monomeric Aurora A, and previous studies have shown that TPX2 binding to the catalytic domain activates Aurora A but not Aurora B. Here we identify G205 in Xenopus Aurora A as a key determinant of both intrinsic activity and regulation by TPX2. Mutation of G205 in Aurora A to N, the equivalent residue in Aurora B, had no effect on autophosphorylation of the T-loop but led to a 10-fold loss of specific activity, whereas mutation of N158 in Aurora B to G caused a 350-fold increase in specific activity. G205 N Aurora A was still activated by TPX2, but protection of pT295 from dephosphorylation by protein phosphatase 1 was abolished. Structural analysis of these effects suggests that the G205 forms a pivot point in the enzyme that results in movement of the N-terminal domain glycine-rich loop closer to the ATP binding site of the enzyme and also moves the C-helix slightly closer to the activation loop. Changes in these positions are comparable to those reported for other protein kinases and demonstrate that phosphorylation of the activation loop alone is not sufficient for enzyme activation. The generation of an activated mutant of Aurora B will be important for studying its role in cell cycle control and tumorigenesis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Xenopus/metabolismo , Sequência de Aminoácidos , Aminoácidos , Animais , Aurora Quinase B , Aurora Quinases , Humanos , Dados de Sequência Molecular , Mutação/genética , Ligação Proteica , Proteínas Serina-Treonina Quinases/química , Estrutura Secundária de Proteína , Proteínas Recombinantes
19.
J Biol Chem ; 280(26): 24339-46, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15860459

RESUMO

Vertebrate oocytes are arrested in G(2) phase of the cell cycle at the prophase border of meiosis I. Progesterone treatment of Xenopus oocytes releases the G(2) block and promotes entry into the M phases of meiosis I and II. Substantial evidence indicates that the release of the G(2) arrest requires a decrease in cAMP and reduced activity of the cAMP-dependent protein kinase (PKAc). It has been reported and we confirm here that microinjection of either wild type or kinase-dead K72R PKAc inhibits progesterone-dependent release of the G(2) arrest with equal potency and that inhibition can be reversed by a second injection of the heat-stable inhibitor of PKAc, PKI. However, a mutant enzyme predicted to be completely kinase-dead from the crystal structure of PKAc, K72H PKAc, was much less inhibitory when carrying additional mutations that block interaction with either type I or type II regulatory subunit. Moreover, inhibition by K72H PKAc was reversed by PKI at a 30-fold lower concentration and with more rapid kinetics compared with wild type PKAc. K72R PKAc was found to have low but detectable activity after incubation in an oocyte extract. These results indicate that inhibition of the progesterone-dependent G(2)/M transition in oocytes after microinjection of dead PKAc reflects either low residual activity or binding to regulatory subunits with a resulting net increase in the level of endogenous wild type PKAc. Consistent with this hypothesis, the induction of mitosis in Xenopus egg extracts by the addition of cyclin B was blocked by wild type PKAc but not by K72H PKAc. The identification of substrates for PKAc that maintain cell cycle arrest in G(2) remains an important goal for future work.


Assuntos
Regulação Enzimológica da Expressão Gênica , Oócitos/metabolismo , Animais , Divisão Celular , Cristalografia por Raios X , AMP Cíclico/metabolismo , Ciclina B/química , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Fase G2 , Cinética , Meiose , Camundongos , Mitose , Oócitos/química , Progesterona/metabolismo , Ligação Proteica , Proteínas Recombinantes/química , Fatores de Tempo , Xenopus laevis
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA