Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 8(61): 103931-103951, 2017 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-29262611

RESUMO

Poly(ADP-ribose) polymerases (PARPs) regulate various aspects of cellular function including mitotic progression. Although PARP inhibitors have been undergoing various clinical trials and the PARP1/2 inhibitor olaparib was approved as monotherapy for BRCA-mutated ovarian cancer, their mode of action in killing tumour cells is not fully understood. We investigated the effect of PARP inhibition on mitosis in cancerous (cervical, ovary, breast and osteosarcoma) and non-cancerous cells by live-cell imaging. The clinically relevant inhibitor olaparib induced strong perturbations in mitosis, including problems with chromosome alignment at the metaphase plate, anaphase delay, and premature loss of cohesion (cohesion fatigue) after a prolonged metaphase arrest, resulting in sister chromatid scattering. PARP1 and PARP2 depletion suppressed the phenotype while PARP2 overexpression enhanced it, suggesting that olaparib-bound PARP1 and PARP2 rather than the lack of catalytic activity causes this phenotype. Olaparib-induced mitotic chromatid scattering was observed in various cancer cell lines with increased protein levels of PARP1 and PARP2, but not in non-cancer or cancer cell lines that expressed lower levels of PARP1 or PARP2. Interestingly, the sister chromatid scattering phenotype occurred only when olaparib was added during the S-phase preceding mitosis, suggesting that PARP1 and PARP2 entrapment at replication forks impairs sister chromatid cohesion. Clinically relevant DNA-damaging agents that impair replication progression such as topoisomerase inhibitors and cisplatin were also found to induce sister chromatid scattering and metaphase plate alignment problems, suggesting that these mitotic phenotypes are a common outcome of replication perturbation.

2.
Elife ; 62017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29106370

RESUMO

The actomyosin ring generates force to ingress the cytokinetic cleavage furrow in animal cells, yet its filament organization and the mechanism of contractility is not well understood. We quantified actin filament order in human cells using fluorescence polarization microscopy and found that cleavage furrow ingression initiates by contraction of an equatorial actin network with randomly oriented filaments. The network subsequently gradually reoriented actin filaments along the cell equator. This strictly depended on myosin II activity, suggesting local network reorganization by mechanical forces. Cortical laser microsurgery revealed that during cytokinesis progression, mechanical tension increased substantially along the direction of the cell equator, while the network contracted laterally along the pole-to-pole axis without a detectable increase in tension. Our data suggest that an asymmetric increase in cortical tension promotes filament reorientation along the cytokinetic cleavage furrow, which might have implications for diverse other biological processes involving actomyosin rings.


Assuntos
Citoesqueleto de Actina/metabolismo , Citocinese , Fenômenos Mecânicos , Miosina Tipo II/metabolismo , Células Cultivadas , Células Epiteliais/fisiologia , Humanos , Microscopia de Fluorescência , Epitélio Pigmentado da Retina/fisiologia
3.
Nat Commun ; 7: 13887, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27976684

RESUMO

Genome integrity relies on precise coordination between DNA replication and chromosome segregation. Whereas replication stress attracted much attention, the consequences of mitotic perturbations for genome integrity are less understood. Here, we knockdown 47 validated mitotic regulators to show that a broad spectrum of mitotic errors correlates with increased DNA breakage in daughter cells. Unexpectedly, we find that only a subset of these correlations are functionally linked. We identify the genuine mitosis-born DNA damage events and sub-classify them according to penetrance of the observed phenotypes. To demonstrate the potential of this resource, we show that DNA breakage after cytokinesis failure is preceded by replication stress, which mounts during consecutive cell cycles and coincides with decreased proliferation. Together, our results provide a resource to gauge the magnitude and dynamics of DNA breakage associated with mitotic aberrations and suggest that replication stress might limit propagation of cells with abnormal karyotypes.


Assuntos
Ciclo Celular , Proliferação de Células , Dano ao DNA/genética , Mitose/genética , Linhagem Celular Tumoral , Citocinese/genética , Quebras de DNA , Técnicas de Silenciamento de Genes , Humanos , Processamento de Imagem Assistida por Computador , Microscopia Confocal , Fenótipo , Imagem com Lapso de Tempo
4.
PLoS One ; 10(7): e0133576, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26201077

RESUMO

RUVBL1 (RuvB-like1) and RUVBL2 (RuvB-like 2) are integral components of multisubunit protein complexes involved in processes ranging from cellular metabolism, transcription and chromatin remodeling to DNA repair. Here, we show that although RUVBL1 and RUVBL2 are known to form heterodimeric complexes in which they stabilize each other, the subunits separate during cytokinesis. In anaphase-to-telophase transition, RUVBL1 localizes to structures of the mitotic spindle apparatus, where it partially co-localizes with polo-like kinase 1 (PLK1). The ability of PLK1 to phosphorylate RUVBL1-but not RUVBL2-in vitro and their physical association in vivo suggest that this kinase differentially regulates the function of the RuvB-like proteins during mitosis. We further show that siRNA-mediated knock-down of RuvB-like proteins causes severe defects in chromosome alignment and segregation. In addition, we show that the ATPase activity of RUVBL1 is indispensable for cell proliferation. Our data thus demonstrate that RUVBL1 is essential for efficient mitosis and proliferation.


Assuntos
Proteínas de Transporte/metabolismo , Segregação de Cromossomos/fisiologia , Citocinese/fisiologia , DNA Helicases/metabolismo , Mitose/fisiologia , ATPases Associadas a Diversas Atividades Celulares , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , DNA Helicases/genética , Reparo do DNA , Células HeLa , Humanos , Fosforilação/fisiologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Fuso Acromático/genética , Fuso Acromático/metabolismo , Quinase 1 Polo-Like
5.
J Pharmacol Exp Ther ; 352(3): 579-89, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25576074

RESUMO

Polo-like kinase 1 (Plk1), a member of the Polo-like kinase family of serine/threonine kinases, is a key regulator of multiple steps in mitosis. Here we report on the pharmacological profile of volasertib, a potent and selective Plk inhibitor, in multiple preclinical models of acute myeloid leukemia (AML) including established cell lines, bone marrow samples from AML patients in short-term culture, and subcutaneous as well as disseminated in vivo models in immune-deficient mice. Our results indicate that volasertib is highly efficacious as a single agent and in combination with established and emerging AML drugs, including the antimetabolite cytarabine, hypomethylating agents (decitabine, azacitidine), and quizartinib, a signal transduction inhibitor targeting FLT3. Collectively, these preclinical data support the use of volasertib as a new therapeutic approach for the treatment of AML patients, and provide a foundation for combination approaches that may further improve and prolong clinical responses.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/enzimologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Pteridinas/uso terapêutico , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Nus , Camundongos SCID , Camundongos Transgênicos , Inibidores de Proteínas Quinases/farmacologia , Pteridinas/farmacologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Quinase 1 Polo-Like
6.
Nat Cell Biol ; 15(11): 1370-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24096243

RESUMO

The mitotic spindle assembly checkpoint (SAC) delays anaphase onset until all chromosomes have attached to both spindle poles. Here, we investigated SAC signalling kinetics in response to acute detachment of individual chromosomes using laser microsurgery. Most detached chromosomes delayed anaphase until they had realigned to the metaphase plate. A substantial fraction of cells, however, entered anaphase in the presence of unaligned chromosomes. We identify two mechanisms by which cells can bypass the SAC: first, single unattached chromosomes inhibit the anaphase-promoting complex/cyclosome (APC/C) less efficiently than a full complement of unattached chromosomes; second, because of the relatively slow kinetics of re-imposing APC/C inhibition during metaphase, cells were unresponsive to chromosome detachment up to several minutes before anaphase onset. Our study defines when cells irreversibly commit to enter anaphase and shows that the SAC signal strength correlates with the number of unattached chromosomes. Detailed knowledge about SAC signalling kinetics is important for understanding the emergence of aneuploidy and the response of cancer cells to chemotherapeutics targeting the mitotic spindle.


Assuntos
Pontos de Checagem da Fase M do Ciclo Celular , Transdução de Sinais , Fuso Acromático , Cromossomos Humanos , Humanos , Cinetocoros
7.
EMBO J ; 32(13): 1886-902, 2013 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-23727888

RESUMO

Nuclei of Xenopus laevis oocytes grow 100 000-fold larger in volume than a typical somatic nucleus and require an unusual intranuclear F-actin scaffold for mechanical stability. We now developed a method for mapping F-actin interactomes and identified a comprehensive set of F-actin binders from the oocyte nuclei. Unexpectedly, the most prominent interactor was a novel kinesin termed NabKin (Nuclear and meiotic actin-bundling Kinesin). NabKin not only binds microtubules but also F-actin structures, such as the intranuclear actin bundles in prophase and the contractile actomyosin ring during cytokinesis. The interaction between NabKin and F-actin is negatively regulated by Importin-ß and is responsive to spatial information provided by RanGTP. Disconnecting NabKin from F-actin during meiosis caused cytokinesis failure and egg polyploidy. We also found actin-bundling activity in Nabkin's somatic paralogue KIF14, which was previously shown to be essential for somatic cell division. Our data are consistent with the notion that NabKin/KIF14 directly link microtubules with F-actin and that such link is essential for cytokinesis.


Assuntos
Actinas/metabolismo , Núcleo Celular/metabolismo , Citocinese/fisiologia , Cinesinas/metabolismo , Meiose/fisiologia , Oócitos/metabolismo , Xenopus laevis/metabolismo , Actomiosina/metabolismo , Animais , Cromatografia de Afinidade , Feminino , Imunofluorescência , Immunoblotting , Microtúbulos/metabolismo , Oócitos/citologia , Faloidina/metabolismo , Ploidias , Proteômica , Proteínas Recombinantes/metabolismo , Espectrometria de Massas em Tandem , Xenopus laevis/crescimento & desenvolvimento
8.
Nat Rev Mol Cell Biol ; 12(8): 469-82, 2011 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-21750572

RESUMO

The mitosis-to-interphase transition involves dramatic cellular reorganization from a state that supports chromosome segregation to a state that complies with all functions of an interphase cell. This process, termed mitotic exit, depends on the removal of mitotic phosphorylations from a broad range of substrates. Mitotic exit regulation involves inactivation of mitotic kinases and activation of counteracting protein phosphatases. The key mitotic exit phosphatase in budding yeast, Cdc14, is now well understood. By contrast, in animal cells, it is now emerging that mitotic exit relies on distinct regulatory networks, including the protein phosphatases PP1 and PP2A.


Assuntos
Mitose/fisiologia , Monoéster Fosfórico Hidrolases/fisiologia , Ciclossomo-Complexo Promotor de Anáfase , Animais , Aurora Quinases , Proteína Quinase CDC2/fisiologia , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/fisiologia , Ciclina B1/fisiologia , Humanos , Interfase/fisiologia , Modelos Biológicos , Neoplasias/patologia , Neoplasias/terapia , Proteína Fosfatase 1/fisiologia , Proteína Fosfatase 2/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Complexos Ubiquitina-Proteína Ligase/fisiologia , Quinase 1 Polo-Like
9.
J Cell Sci ; 124(Pt 9): 1571-80, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21486945

RESUMO

During exit from mitosis in Xenopus laevis egg extracts, the AAA+ ATPase Cdc48/p97 (also known as VCP in vertebrates) and its adapter Ufd1-Npl4 remove the kinase Aurora B from chromatin to allow nucleus formation. Here, we show that in HeLa cells Ufd1-Npl4 already antagonizes Aurora B on chromosomes during earlier mitotic stages and that this is crucial for proper chromosome segregation. Depletion of Ufd1-Npl4 by small interfering RNA (siRNA) caused chromosome alignment and anaphase defects resulting in missegregated chromosomes and multi-lobed nuclei. Ufd1-Npl4 depletion also led to increased levels of Aurora B on prometaphase and metaphase chromosomes. This increase was associated with higher Aurora B activity, as evidenced by the partial resistance of CENP-A phosphorylation to the Aurora B inhibitor hesperadin. Furthermore, low concentrations of hesperadin partially rescued chromosome alignment in Ufd1-depleted cells, whereas, conversely, Ufd1-depletion partially restored congression in the presence of hesperadin. These data establish Cdc48/p97-Ufd1-Npl4 as a crucial negative regulator of Aurora B early in mitosis of human somatic cells and suggest that the activity of Aurora B on chromosomes needs to be restrained to ensure faithful chromosome segregation.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Segregação de Cromossomos/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Adenosina Trifosfatases/genética , Animais , Aurora Quinase B , Aurora Quinases , Western Blotting , Proteínas de Ciclo Celular/genética , Segregação de Cromossomos/genética , Técnica Indireta de Fluorescência para Anticorpo , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Mitose/genética , Mitose/fisiologia , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas/genética , RNA Interferente Pequeno , Proteína com Valosina
10.
Science ; 331(6024): 1616-20, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21310966

RESUMO

After partitioning of cytoplasmic contents by cleavage furrow ingression, animal cells remain connected by an intercellular bridge, which subsequently splits by abscission. Here, we examined intermediate stages of abscission in human cells by using live imaging, three-dimensional structured illumination microscopy, and electron tomography. We identified helices of 17-nanometer-diameter filaments, which narrowed the cortex of the intercellular bridge to a single stalk. The endosomal sorting complex required for transport (ESCRT)-III co-localized with constriction zones and was required for assembly of 17-nanometer-diameter filaments. Simultaneous spastin-mediated removal of underlying microtubules enabled full constriction at the abscission site. The identification of contractile filament helices at the intercellular bridge has broad implications for the understanding of cell division and of ESCRT-III-mediated fission of large membrane structures.


Assuntos
Divisão Celular , Complexos Endossomais de Distribuição Requeridos para Transporte/química , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Actinas/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Tomografia com Microscopia Eletrônica , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Células HeLa , Humanos , Imageamento Tridimensional , Microscopia Eletrônica , Proteínas Nucleares/metabolismo , Conformação Proteica , Multimerização Proteica , Interferência de RNA , Espastina
11.
EMBO J ; 29(24): 4210-22, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21113133

RESUMO

Cullin 4 (Cul4)-based ubiquitin ligases emerged as critical regulators of DNA replication and repair. Over 50 Cul4-specific adaptors (DNA damage-binding 1 (Ddb1)-Cul4-associated factors; DCAFs) have been identified and are thought to assemble functionally distinct Cul4 complexes. Using a live-cell imaging-based RNAi screen, we analysed the function of DCAFs and Cul4-linked proteins, and identified specific subsets required for progression through G1 and S phase. We discovered C6orf167/Mms22-like protein (Mms22L) as a putative human orthologue of budding yeast Mms22, which, together with cullin Rtt101, regulates genome stability by promoting DNA replication through natural pause sites and damaged templates. Loss of Mms22L function in human cells results in S phase-dependent genomic instability characterised by spontaneous double-strand breaks and DNA damage checkpoint activation. Unlike yeast Mms22, human Mms22L does not stably bind to Cul4, but is degraded in a Cul4-dependent manner and upon replication stress. Mms22L physically and functionally interacts with the scaffold-like protein Nfkbil2 that co-purifies with histones, several chromatin remodelling and DNA replication/repair factors. Together, our results strongly suggest that the Mms22L-Nfkbil2 complex contributes to genome stability by regulating the chromatin state at stalled replication forks.


Assuntos
Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/antagonistas & inibidores , Técnicas de Silenciamento de Genes/métodos , Instabilidade Genômica , Humanos , Programas de Rastreamento/métodos , Proteínas Nucleares/antagonistas & inibidores , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
12.
J Cell Biol ; 189(6): 945-54, 2010 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-20530212

RESUMO

Posttranslational glutamylation of tubulin is present on selected subsets of microtubules in cells. Although the modification is expected to contribute to the spatial and temporal organization of the cytoskeleton, hardly anything is known about its functional relevance. Here we demonstrate that glutamylation, and in particular the generation of long glutamate side chains, promotes the severing of microtubules. In human cells, the generation of long side chains induces spastin-dependent microtubule disassembly and, consistently, only microtubules modified by long glutamate side chains are efficiently severed by spastin in vitro. Our study reveals a novel control mechanism for microtubule mass and stability, which is of fundamental importance to cellular physiology and might have implications for diseases related to microtubule severing.


Assuntos
Adenosina Trifosfatases/metabolismo , Ácido Glutâmico , Microtúbulos/metabolismo , Processamento de Proteína Pós-Traducional , Tubulina (Proteína)/metabolismo , Adenosina Trifosfatases/genética , Animais , Citoesqueleto/metabolismo , Ácido Glutâmico/química , Ácido Glutâmico/metabolismo , Células HeLa , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Katanina , Camundongos , Peptídeo Sintases/genética , Peptídeo Sintases/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Espastina , Tubulina (Proteína)/química
13.
J Cell Sci ; 123(Pt 9): 1395-400, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20356927

RESUMO

Germline mutations in the tumor-suppressor gene BRCA2 predispose to breast and ovarian cancer. BRCA2 plays a well-established role in maintaining genome stability by regulating homologous recombination. BRCA2 has more recently been implicated in cytokinesis, the final step of cell division, but the molecular basis for this remains unknown. We have used time-lapse microscopy, recently developed cytokinesis assays and BAC recombineering (bacterial artificial chromosome recombinogenic engineering) to investigate the function and localization of BRCA2 during cell division. Our analysis suggests that BRCA2 does not regulate cytokinesis in human cells. Thus, cytokinesis defects are unlikely to contribute to chromosomal instability and tumorigenesis in BRCA2-related cancers.


Assuntos
Proteína BRCA2/metabolismo , Citocinese , Proteínas Reguladoras de Apoptose , Núcleo Celular/metabolismo , Cromossomos Artificiais Bacterianos/metabolismo , Marcação de Genes , Células HeLa , Humanos , Microtúbulos/metabolismo , RNA Interferente Pequeno/metabolismo , Rad51 Recombinase/metabolismo , Fuso Acromático/metabolismo , Fatores de Tempo , Transfecção
14.
J Cell Biol ; 185(5): 859-74, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19468067

RESUMO

Mitotic spindle formation and chromosome segregation depend critically on kinetochore-microtubule (KT-MT) interactions. A new protein, termed Spindly in Drosophila and SPDL-1 in C. elegans, was recently shown to regulate KT localization of dynein, but depletion phenotypes revealed striking differences, suggesting evolutionarily diverse roles of mitotic dynein. By characterizing the function of Spindly in human cells, we identify specific functions for KT dynein. We show that localization of human Spindly (hSpindly) to KTs is controlled by the Rod/Zw10/Zwilch (RZZ) complex and Aurora B. hSpindly depletion results in reduced inter-KT tension, unstable KT fibers, an extensive prometaphase delay, and severe chromosome misalignment. Moreover, depletion of hSpindly induces a striking spindle rotation, which can be rescued by co-depletion of dynein. However, in contrast to Drosophila, hSpindly depletion does not abolish the removal of MAD2 and ZW10 from KTs. Collectively, our data reveal hSpindly-mediated dynein functions and highlight a critical role of KT dynein in spindle orientation.


Assuntos
Dineínas/metabolismo , Cinetocoros/metabolismo , Mitose , Fuso Acromático/metabolismo , Aurora Quinase B , Aurora Quinases , Cromossomos Humanos/metabolismo , Cromossomos Humanos/ultraestrutura , Proteínas do Citoesqueleto , Complexo Dinactina , Células HeLa , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Paclitaxel/farmacologia , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais , Fuso Acromático/ultraestrutura
16.
Cell ; 136(3): 473-84, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19203582

RESUMO

Genomic abnormalities are often seen in tumor cells, and tetraploidization, which results from failures during cytokinesis, is presumed to be an early step in cancer formation. Here, we report a cell division control mechanism that prevents tetraploidization in human cells with perturbed chromosome segregation. First, we found that Aurora B inactivation promotes completion of cytokinesis by abscission. Chromosome bridges sustained Aurora B activity to posttelophase stages and thereby delayed abscission at stabilized intercellular canals. This was essential to suppress tetraploidization by furrow regression in a pathway further involving the phosphorylation of mitotic kinesin-like protein 1 (Mklp1). We propose that Aurora B is part of a sensor that responds to unsegregated chromatin at the cleavage site. Our study provides evidence that in human cells abscission is coordinated with the completion of chromosome segregation to protect against tetraploidization by furrow regression.


Assuntos
Segregação de Cromossomos , Citocinese , Ploidias , Proteínas Serina-Treonina Quinases/metabolismo , Aurora Quinase B , Aurora Quinases , Divisão Celular , Células HeLa , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA