Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Cell Sci ; 127(Pt 11): 2493-506, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24695856

RESUMO

During mitotic entry, centrosomes separate to establish the bipolar spindle. Delays in centrosome separation can perturb chromosome segregation and promote genetic instability. However, interphase centrosomes are physically tethered by a proteinaceous linker composed of C-Nap1 (also known as CEP250) and the filamentous protein rootletin. Linker disassembly occurs at the onset of mitosis in a process known as centrosome disjunction and is triggered by the Nek2-dependent phosphorylation of C-Nap1. However, the mechanistic consequences of C-Nap1 phosphorylation are unknown. Here, we demonstrate that Nek2 phosphorylates multiple residues within the C-terminal domain of C-Nap1 and, collectively, these phosphorylation events lead to loss of oligomerization and centrosome association. Mutations in non-phosphorylatable residues that make the domain more acidic are sufficient to release C-Nap1 from the centrosome, suggesting that it is an increase in overall negative charge that is required for this process. Importantly, phosphorylation of C-Nap1 also perturbs interaction with the core centriolar protein, Cep135, and interaction of endogenous C-Nap1 and Cep135 proteins is specifically lost in mitosis. We therefore propose that multisite phosphorylation of C-Nap1 by Nek2 perturbs both oligomerization and Cep135 interaction, and this precipitates centrosome disjunction at the onset of mitosis.


Assuntos
Autoantígenos/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centríolos/metabolismo , Centrossomo/fisiologia , Fuso Acromático/metabolismo , Autoantígenos/genética , Proteínas de Ciclo Celular/genética , Segregação de Cromossomos/genética , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Instabilidade Genômica , Células HeLa , Humanos , Mitose , Mutação/genética , Quinases Relacionadas a NIMA , Fosforilação , Ligação Proteica/genética , Engenharia de Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/genética
2.
J Cell Sci ; 125(Pt 22): 5353-68, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22956538

RESUMO

Centrosome duplication is licensed by the disengagement, or 'uncoupling', of centrioles during late mitosis. However, arrest of cells in G2 can trigger premature centriole disengagement. Here, we show that premature disengagement results from untimely activation of the anaphase-promoting complex (APC/C), leading to securin degradation and release of active separase. Although APC/C activation during G2 arrest is dependent on polo-like kinase 1 (Plk1)-mediated degradation of the APC/C inhibitor, early mitotic inhibitor 1 (Emi1), Plk1 also has a second APC/C-independent role in promoting disengagement. Importantly, APC/C and Plk1 activity also stimulates centriole disengagement in response to hydroxyurea or DNA damage-induced cell-cycle arrest and this leads to centrosome amplification. However, the reduplication of disengaged centrioles is dependent on cyclin-dependent kinase 2 (Cdk2) activity and Cdk2 activation coincides with a subsequent inactivation of the APC/C and re-accumulation of cyclin A. Although release from these arrests leads to mitotic entry, the presence of disengaged and/or amplified centrosomes results in the formation of abnormal mitotic spindles that lead to chromosome mis-segregation. Thus, oscillation of APC/C activity during cell cycle arrest promotes both centrosome amplification and genome instability.


Assuntos
Pontos de Checagem do Ciclo Celular , Centrossomo/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Proteínas de Ciclo Celular/metabolismo , Centríolos/efeitos dos fármacos , Centríolos/metabolismo , Centríolos/efeitos da radiação , Centrossomo/efeitos dos fármacos , Centrossomo/efeitos da radiação , Endopeptidases/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/efeitos da radiação , Células HeLa , Humanos , Hidroxiureia/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Radiação Ionizante , Separase , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo , Fuso Acromático/efeitos da radiação , Quinase 1 Polo-Like
3.
J Med Chem ; 55(7): 3228-41, 2012 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-22404346
4.
Nat Cell Biol ; 12(12): 1166-76, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21076410

RESUMO

During interphase, centrosomes are held together by a proteinaceous linker that connects the proximal ends of the mother and daughter centriole. This linker is disassembled at the onset of mitosis in a process known as centrosome disjunction, thereby facilitating centrosome separation and bipolar spindle formation. The NIMA (never in mitosis A)-related kinase Nek2A is implicated in disconnecting the centrosomes through disjoining the linker proteins C-Nap1 and rootletin. However, the mechanisms controlling centrosome disjunction remain poorly understood. Here, we report that two Hippo pathway components, the mammalian sterile 20-like kinase 2 (Mst2) and the scaffold protein Salvador (hSav1), directly interact with Nek2A and regulate its ability to localize to centrosomes, and phosphorylate C-Nap1 and rootletin. Furthermore, we demonstrate that the hSav1-Mst2-Nek2A centrosome disjunction pathway becomes essential for bipolar spindle formation on partial inhibition of the kinesin-5 Eg5. We propose that hSav1-Mst2-Nek2A and Eg5 have distinct, but complementary functions, in centrosome disjunction.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Humanos , Interfase , Dados de Sequência Molecular , Quinases Relacionadas a NIMA , Alinhamento de Sequência , Serina-Treonina Quinase 3
5.
J Mol Biol ; 386(2): 476-85, 2009 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19124027

RESUMO

The Nek family of serine/threonine kinases regulates centrosome and cilia function; in addition, several of its members are potential targets for drug discovery. Nek2 is dimeric, is cell cycle regulated and functions in the separation of centrosomes at G2/M. Here, we report the crystal structures of wild-type human Nek2 kinase domain bound to ADP at 1.55-A resolution and T175A mutant in apo form as well as that bound to a non-hydrolyzable ATP analog. These show that regions of the Nek2 structure around the nucleotide-binding site can adopt several different but well-defined conformations. None of the conformations was the same as that observed for the previously reported inhibitor-bound structure, and the two nucleotides stabilized two conformations. The structures suggest mechanisms for the auto-inhibition of Nek2 that we have tested by mutagenesis. Comparison of the structures with Aurora-A and Cdk2 gives insight into the structural mechanism of Nek2 activation. The production of specific inhibitors that target individual kinases of the human genome is an urgent challenge in drug discovery, and Nek2 is especially promising as a cancer target. We not only identify potential challenges to the task of producing Nek2 inhibitors but also propose that the conformational variability provides an opportunity for the design of Nek2 selective inhibitors because one of the conformations may provide a unique target.


Assuntos
Proteínas Serina-Treonina Quinases/química , Sítios de Ligação , Cristalografia por Raios X , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Mutação de Sentido Incorreto , Quinases Relacionadas a NIMA , Conformação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
6.
Mol Cell ; 32(4): 576-83, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-19026787

RESUMO

The Fizzy/Cdc20 family of proteins are essential activators of the anaphase-promoting complex/cyclosome (APC/C), a multisubunit E3 ubiquitin ligase. However, apart from the well-established role of the C-terminal WD40 domain in substrate recognition, the precise roles of the activators remain elusive. Here we show that Nek2A, which directly binds the APC/C, can be ubiquitylated and destroyed in Fizzy/Cdc20-depleted Xenopus egg extracts when only the N-terminal domain of Fizzy/Cdc20 (N-Cdc20) is added. This activity is dependent upon the C box and is conserved in the alternative activator, Fizzy-related/Cdh1. In contrast, canonical substrates such as cyclin B and securin require both the N-terminal and WD40 domains, unless N-Cdc20 is fused to substrates when the WD40 domain becomes dispensable. Furthermore, in Cdc20-depleted cells, N-Cdc20 can facilitate Nek2A destruction in a C box-dependent manner. Our results reveal a role for the N-terminal domain of the Fizzy/Cdc20 family of activators in triggering substrate ubiquitylation by the APC/C.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Proteínas de Xenopus/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Animais , Proteínas Cdc20 , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Modelos Biológicos , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas/genética , Especificidade por Substrato , Ubiquitinação , Xenopus/genética , Xenopus/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/genética
7.
J Biol Chem ; 282(36): 26431-40, 2007 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-17626005

RESUMO

Nek2 is a cell cycle-regulated serine/threonine protein kinase that is up-regulated in human cancers. Functionally, it is implicated in control of centrosome separation and bipolar spindle formation in mitotic cells and chromatin condensation in meiotic cells. Two major splice variants have been described in vertebrates, Nek2A and Nek2B, that differ in their non-catalytic C termini. Recently, a third splice variant, Nek2C, was identified that lacks an eight-amino acid internal sequence within the C-terminal domain of Nek2A. This excision occurs at the same position as the Nek2A/Nek2B splice point. As predicted from their high degree of similarity, we show here that Nek2C shares many properties with Nek2A including kinase activity, dimerization, protein phosphatase 1 interaction, mitotic degradation, microtubule binding, and centrosome localization. Unexpectedly, though, the non-centrosomal pool of protein exhibits a marked difference in distribution for the three splice variants. Nek2C is mainly nuclear, Nek2B is mainly cytoplasmic, and Nek2A is evenly distributed within nuclei and cytoplasm. Mutagenesis experiments revealed a functional bipartite nuclear localization sequence (NLS) that spans the splice site leading to Nek2C having a strong NLS, Nek2A having a weak NLS, and Nek2B having no NLS. Finally, we identified a 28-kDa protein in nuclear extracts as a potential novel substrate of Nek2. Thus, alternative splicing provides an unusual mechanism for modulating Nek2 localization, enabling it to have both nuclear and cytoplasmic functions.


Assuntos
Processamento Alternativo , Núcleo Celular/metabolismo , Centrossomo/metabolismo , Montagem e Desmontagem da Cromatina , Proteínas Serina-Treonina Quinases/metabolismo , Fuso Acromático/metabolismo , Transporte Ativo do Núcleo Celular/genética , Processamento Alternativo/genética , Núcleo Celular/patologia , Centrossomo/patologia , Montagem e Desmontagem da Cromatina/genética , Dimerização , Células HeLa , Humanos , Mitose/genética , Mutação , Quinases Relacionadas a NIMA , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Sinais de Localização Nuclear/genética , Fosfoproteínas Fosfatases/metabolismo , Ligação Proteica/genética , Proteína Fosfatase 1 , Proteínas Serina-Treonina Quinases/genética , Sítios de Splice de RNA/genética , Fuso Acromático/patologia , Regulação para Cima/genética
8.
J Biol Chem ; 282(9): 6833-42, 2007 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-17197699

RESUMO

The dimeric Ser/Thr kinase Nek2 regulates centrosome cohesion and separation through phosphorylation of structural components of the centrosome, and aberrant regulation of Nek2 activity can lead to aneuploid defects characteristic of cancer cells. Mutational analysis of autophosphorylation sites within the kinase domain identified by mass spectrometry shows a complex pattern of positive and negative regulatory effects on kinase activity that are correlated with effects on centrosomal splitting efficiency in vivo. The 2.2-A resolution x-ray structure of the Nek2 kinase domain in complex with a pyrrole-indolinone inhibitor reveals an inhibitory helical motif within the activation loop. This helix presents a steric barrier to formation of the active enzyme and generates a surface that may be exploitable in the design of specific inhibitors that selectively target the inactive state. Comparison of this "auto-inhibitory" conformation with similar arrangements in cyclin-dependent kinase 2 and epidermal growth factor receptor kinase suggests a role for dimerization-dependent allosteric regulation that combines with autophosphorylation and protein phosphatase 1c phosphatase activity to generate the precise spatial and temporal control required for Nek2 function in centrosomal maturation.


Assuntos
Regulação Alostérica , Centrossomo/enzimologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/química , Sítios de Ligação , Cristalografia por Raios X , Análise Mutacional de DNA , Dimerização , Humanos , Espectrometria de Massas , Quinases Relacionadas a NIMA , Fosforilação
9.
Dev Cell ; 11(4): 431-2, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17011480

RESUMO

The kinase Plk1 plays multiple roles in regulating mitotic progression, including stabilization of spindle poles, but its substrates are largely unknown. A new study by Yamamoto and coworkers has identified a centrosomal protein, Kizuna (Kiz), as a mitotic substrate of Plk1 (Oshimori et al., 2006). Phosphorylation of Kiz ensures the integrity of spindle poles in the face of severe pulling forces exerted by the chromosome-attached spindle microtubules.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fuso Acromático/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular/química , Centrossomo/metabolismo , Humanos , Microtúbulos/fisiologia , Mitose/fisiologia , Modelos Biológicos , Dados de Sequência Molecular , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Proto-Oncogênicas/química , Fuso Acromático/fisiologia , Treonina/metabolismo , Quinase 1 Polo-Like
10.
Mol Cell Biol ; 25(4): 1309-24, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15684383

RESUMO

Mitotic entry requires a major reorganization of the microtubule cytoskeleton. Nlp, a centrosomal protein that binds gamma-tubulin, is a G(2)/M target of the Plk1 protein kinase. Here, we show that human Nlp and its Xenopus homologue, X-Nlp, are also phosphorylated by the cell cycle-regulated Nek2 kinase. X-Nlp is a 213-kDa mother centriole-specific protein, implicating it in microtubule anchoring. Although constant in abundance throughout the cell cycle, it is displaced from centrosomes upon mitotic entry. Overexpression of active Nek2 or Plk1 causes premature displacement of Nlp from interphase centrosomes. Active Nek2 is also capable of phosphorylating and displacing a mutant form of Nlp that lacks Plk1 phosphorylation sites. Importantly, kinase-inactive Nek2 interferes with Plk1-induced displacement of Nlp from interphase centrosomes and displacement of endogenous Nlp from mitotic spindle poles, while active Nek2 stimulates Plk1 phosphorylation of Nlp in vitro. Unlike Plk1, Nek2 does not prevent association of Nlp with gamma-tubulin. Together, these results provide the first example of a protein involved in microtubule organization that is coordinately regulated at the G(2)/M transition by two centrosomal kinases. We also propose that phosphorylation by Nek2 may prime Nlp for phosphorylation by Plk1.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centríolos/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Xenopus/metabolismo , Sequência de Aminoácidos , Animais , Ciclo Celular/fisiologia , Humanos , Rim/metabolismo , Microtúbulos/metabolismo , Dados de Sequência Molecular , Quinases Relacionadas a NIMA , Fosforilação , Proteínas Proto-Oncogênicas , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas , Xenopus laevis , Quinase 1 Polo-Like
11.
Dev Biol ; 265(2): 384-98, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14732400

RESUMO

Pronuclear migration and formation of the first mitotic spindle depend upon assembly of a functional zygotic centrosome. For most animals, this involves both paternal and maternal contributions as sperm basal bodies are converted into centrosomes competent for microtubule nucleation through recruitment of egg proteins. Nek2B is a vertebrate NIMA-related protein kinase required for centrosome assembly, as its depletion from egg extracts delays microtubule aster formation from sperm basal bodies. Using Xenopus as a model system, we now show that protein expression of Nek2B begins during mid-oogenesis and increases further upon oocyte maturation. This is regulated, at least in part, at the level of protein translation. Nek2B protein is weakly phosphorylated in mitotic egg extracts but its recruitment to the sperm basal body, which occurs independently of its kinase activity, stimulates its phosphorylation, possibly through sequestration from a phosphatase present in mitotic egg cytoplasm. Importantly, although Nek2B is not required to organize acentrosomal microtubule asters, we show that addition of either active or kinase-dead recombinant Nek2B can restore centrosome assembly in a dose-dependent manner to a depleted extract. These results support a model in which maternal Nek2B acts to promote assembly of a functional zygotic centrosome in a kinase-independent manner.


Assuntos
Centrossomo/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Xenopus , Zigoto/enzimologia , Animais , Centrossomo/metabolismo , Citoplasma/enzimologia , Feminino , Isoenzimas/metabolismo , Masculino , Microtúbulos/enzimologia , Oócitos/enzimologia , Oogênese/fisiologia , Óvulo/enzimologia , Espermatozoides/enzimologia , Xenopus laevis , Zigoto/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA