Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
Intervalo de ano de publicação
1.
J Cell Sci ; 132(20)2019 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-31519809

RESUMO

AMP-activated protein kinase (AMPK), a heterotrimeric serine/threonine kinase and cellular metabolic sensor, has been found to regulate cell cycle checkpoints in cancer cells in response to energetic stress, to harmonize proliferation with energy availability. Despite AMPK's emergent association with the cell cycle, it still has not been fully delineated how AMPK is regulated by upstream signaling pathways during mitosis. We report, for the first time, direct CDK1 phosphorylation of both the catalytic α1 and α2 subunits, as well as the ß1 regulatory subunit, of AMPK in mitosis. We found that AMPK-knockout U2OS osteosarcoma cells have reduced mitotic indexes and that CDK1 phosphorylation-null AMPK is unable to rescue the phenotype, demonstrating a role for CDK1 regulation of mitotic entry through AMPK. Our results also denote a vital role for AMPK in promoting proper chromosomal alignment, as loss of AMPK activity leads to misaligned chromosomes and concomitant metaphase delay. Importantly, AMPK expression and activity was found to be critical for paclitaxel chemosensitivity in breast cancer cells and positively correlated with relapse-free survival in systemically treated breast cancer patients.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteína Quinase CDC2/metabolismo , Cromossomos Humanos/metabolismo , Metáfase , Proteínas Quinases Ativadas por AMP/genética , Proteína Quinase CDC2/genética , Cromossomos Humanos/genética , Células HEK293 , Células HeLa , Humanos , Células MCF-7
2.
Cell Signal ; 69: 109546, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31981797

RESUMO

Protein kinase N1 (PKN1) is a member of the protein kinase C superfamily. Aberrations of PKN1 kinase activity are involved in several human pathological processes, including cancer. We found that PKN family proteins (PKN1/2/3) are phosphorylated in response to antitubulin drug-induced mitotic arrest. We identified cyclin-dependent kinase 1 (CDK1) as the corresponding kinase for PKN protein phosphorylation. CDK1 phosphorylates PKN1 at S533, S537, S562, and S916 in vitro and in cells during drug-induced mitotic arrest. Immunofluorescence staining further confirmed that PKN1 phosphorylation occurs during normal mitosis in a CDK1-dependent manner. Knockdown of PKN1 significantly inhibited anchorage-independent growth and migration without affecting proliferation in multiple cancer cell lines. We further showed that mitotic phosphorylation is essential for PKN1's oncogenic function, as the non-phosphorylatable mutant PKN1-4A failed to rescue anchorage-independent growth and migration in PKN1-knockdown cells. Thus, our findings reveal a novel regulatory mechanism for PKN1 in mitosis and its role in tumorigenesis.


Assuntos
Proteína Quinase CDC2/metabolismo , Carcinogênese/metabolismo , Proteína Quinase C/metabolismo , Movimento Celular , Proliferação de Células , Células HEK293 , Células HeLa , Humanos , Fosforilação
3.
FEBS J ; 286(5): 963-974, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30600590

RESUMO

LIM domains containing 1 (LIMD1) is a member of the Zyxin family proteins and functions as a tumor suppressor in lung cancer. LIMD1 has been shown to regulate Hippo-YAP signaling activity. Here, we report a novel regulatory mechanism for LIMD1. We found that cyclin-dependent kinase 1 (CDK1) and c-Jun NH2-terminal kinases 1/2 (JNK1/2) phosphorylate LIMD1 in vitro and in cells during anti-tubulin drug-induced mitotic arrest. Phosphorylation also occurs during normal mitosis. S272, S277, S421, and S424 were identified as the main phosphorylation sites in LIMD1. Deletion of LIMD1 resulted in a shortened mitotic cell cycle and phosphorylation of LIMD1 is required for proper mitotic progression. We further showed that the phosphorylation-deficient mutant LIMD1-4A is less active in suppressing cell proliferation, anchorage-independent growth, cell migration, and invasion in lung cancer cells. Together, our findings suggest that LIMD1 is a key regulator of mitotic progression, and that dysregulation of LIMD1 contributes to tumorigenesis.


Assuntos
Genes Supressores de Tumor , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/metabolismo , Mitose/fisiologia , Proteína Quinase CDC2/metabolismo , Adesão Celular , Proliferação de Células/fisiologia , Transformação Celular Neoplásica , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas com Domínio LIM/fisiologia , Mitose/efeitos dos fármacos , Invasividade Neoplásica , Fosforilação , Moduladores de Tubulina/farmacologia
4.
Cell Signal ; 52: 137-146, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30223016

RESUMO

YES is a member of the SRC family kinase (SFK) group of non-receptor tyrosine kinases, which are implicated in multiple key cellular processes involved in oncogenesis. Antitubulin agents have been widely used as chemotherapeutics for cancer patients and these drugs arrest cells in mitosis, leading to subsequent cell death. In the present study, we define a mechanism for phospho-regulation of YES that is critical for its role in response to antitubulin agents. Specifically, we found that YES is phosphorylated at multiple sites on its N-terminal unique domain by the cell cycle kinase CDK1 during antitubulin drug-induced mitotic arrest. Phosphorylation of YES occurs during normal mitosis. Deletion of YES causes arrest in prometaphase and polyploidy in a p53-independent manner. We further show that YES regulates antitubulin chemosensitivity. Importantly, mitotic phosphorylation is essential for these effects. In support of our findings, we found that YES expression is high in recurrent ovarian cancer patients. Finally, through expression profiling, we documented that YES phosphorylation affects expression of multiple cell cycle regulators. Collectively, our results reveal a previously unrecognized mechanism for controlling the activity of YES during antitubulin chemotherapeutic treatment and suggest YES as a potential target for the treatment of antitubulin-resistant cancer.


Assuntos
Apoptose/efeitos dos fármacos , Proteína Quinase CDC2/metabolismo , Resistencia a Medicamentos Antineoplásicos , Mitose/efeitos dos fármacos , Nocodazol/farmacologia , Paclitaxel/farmacologia , Proteínas Proto-Oncogênicas c-yes/metabolismo , Moduladores de Tubulina/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Nocodazol/uso terapêutico , Paclitaxel/uso terapêutico , Fosforilação , Proteínas Proto-Oncogênicas c-yes/genética , Moduladores de Tubulina/uso terapêutico
5.
Cell Signal ; 39: 74-83, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28780319

RESUMO

PDZ-binding kinase (PBK) plays a major role in proliferation and in safeguarding mitotic fidelity in cancer cells. Frequently upregulated in many cancers, PBK drives tumorigenesis and metastasis. PBK has been shown to be phosphorylated in mitosis by cyclin-dependent kinase 1 (CDK1)/cyclin B, however, no studies have been done examining PBK mitotic phosphorylation in oncogenesis. Additionally to the previously identified Threonine-9 phosphorylation, we found that Threonine-24, Serine-32, and Serine-59 of PBK are also phosphorylated. PBK is phosphorylated in vitro and in cells by CDK1 during antimitotic drug-induced mitotic arrest and in normal mitosis. We demonstrated that mitotic phosphorylation of Threonine-9 is involved in cytokinesis. The non-phosphorylatable mutant PBK-T9A augments tumorigenesis to a greater extent than wild type PBK in breast cancer cells, suggesting that PBK mitotic phosphorylation inhibits its tumor promoting activity. The PBK-T9A mutant also transforms and increases the proliferation of immortalized breast epithelial cells. Collectively, this study reveals that CDK1-mediated mitotic phosphorylation of PBK is involved in cytokinesis and inhibits its oncogenic activity.


Assuntos
Proteína Quinase CDC2/metabolismo , Carcinogênese/metabolismo , Citocinese , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mitose , Proteína Quinase CDC2/genética , Linhagem Celular Tumoral , Proliferação de Células , Células HEK293 , Células HeLa , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Mutação , Fosforilação , Serina/metabolismo , Treonina/metabolismo
6.
Cell Signal ; 28(12): 1826-1832, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27566175

RESUMO

Mammalian sterile 20-like kinase 1/2 (MST1/2) are core tumor suppressors in the Hippo signaling pathway. MST1/2 have been shown to regulate mitotic progression. Here, we report a novel mechanism for phospho-regulation of MST2 in mitosis and its biological significance in cancer. We found that the mitotic kinase cyclin-dependent kinase 1 (CDK1) phosphorylates MST2 in vitro and in vivo at serine 385 during antimitotic drug-induced G2/M phase arrest. This phosphorylation occurs transiently during unperturbed mitosis. Mitotic phosphorylation of MST2 does not affect its kinase activity or Hippo-YAP signaling. We further showed that mitotic phosphorylation-deficient mutant MST2-S385A possesses higher activity in suppressing cell proliferation and anchorage-independent growth in vitro and tumorigenesis in vivo. Together, our findings reveal a novel layer of regulation for MST2 in mitosis and its role in tumorigenesis.


Assuntos
Mitose , Fosfosserina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Antimitóticos/farmacologia , Proteína Quinase CDC2/metabolismo , Carcinogênese/efeitos dos fármacos , Carcinogênese/metabolismo , Carcinogênese/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina B/metabolismo , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos Nus , Mitose/efeitos dos fármacos , Proteínas Mutantes/metabolismo , Fosforilação/efeitos dos fármacos , Serina-Treonina Quinase 3 , Transdução de Sinais/efeitos dos fármacos
7.
Oncotarget ; 6(34): 36019-31, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26440309

RESUMO

The transcriptional co-activator Yes-associated protein, YAP, is a main effector in the Hippo tumor suppressor pathway. We recently defined a mechanism for positive regulation of YAP through CDK1-mediated mitotic phosphorylation. Here, we show that active YAP promotes pancreatic cancer cell migration, invasion and anchorage-independent growth in a mitotic phosphorylation-dependent manner. Mitotic phosphorylation is essential for YAP-driven tumorigenesis in animals. YAP reduction significantly impairs cell migration and invasion. Immunohistochemistry shows significant upregulation and nuclear localization of YAP in metastases when compared with primary tumors and normal tissue in human. Mitotic phosphorylation of YAP controls a unique transcriptional program in pancreatic cells. Expression profiles reveal LPAR3 (lysophosphatidic acid receptor 3) as a mediator for mitotic phosphorylation-driven pancreatic cell motility and invasion. Together, this work identifies YAP as a novel regulator of pancreatic cancer cell motility, invasion and metastasis, and as a potential therapeutic target for invasive pancreatic cancer.


Assuntos
Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/patologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Carcinogênese , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Células HEK293 , Células HeLa , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosforilação , Receptores de Ácidos Lisofosfatídicos/genética , Transdução de Sinais , Fatores de Transcrição/genética , Transfecção
8.
Oncotarget ; 6(31): 31399-412, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26375055

RESUMO

The transcriptional co-activator with PDZ-binding motif (TAZ) is a downstream effector of the Hippo tumor suppressor pathway, which plays important roles in cancer and stem cell biology. Hippo signaling inactivates TAZ through phosphorylation (mainly at S89). In the current study, we define a new layer of regulation of TAZ activity that is critical for its oncogenic function. We found that TAZ is phosphorylated in vitro and in vivo by the mitotic kinase CDK1 at S90, S105, T326, and T346 during the G2/M phase of the cell cycle. Interestingly, mitotic phosphorylation inactivates TAZ oncogenic activity, as the non-phosphorylatable mutant (TAZ-S89A/S90A/S105A/T326A/T346A, TAZ-5A) possesses higher activity in epithelial-mesenchymal transition, anchorage-independent growth, cell migration, and invasion when compared to the TAZ-S89A mutant. Accordingly, TAZ-5A has higher transcriptional activity compared to the TAZ-S89A mutant. Finally, we show that TAZ-S89A or TAZ-5A (to a greater extent) was sufficient to induce spindle and centrosome defects, and chromosome misalignment/missegregation in immortalized epithelial cells. Together, our results reveal a previously unrecognized connection between TAZ oncogenicity and mitotic phospho-regulation.


Assuntos
Transformação Celular Neoplásica/patologia , Quinases Ciclina-Dependentes/metabolismo , Mitose/fisiologia , Neoplasias/prevenção & controle , Fatores de Transcrição/metabolismo , Aciltransferases , Western Blotting , Proteína Quinase CDC2 , Movimento Celular , Células Cultivadas , Transição Epitelial-Mesenquimal , Imunofluorescência , Células HEK293 , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Neoplasias/patologia , Fosforilação , Transdução de Sinais
9.
Nucleus ; 5(5): 462-73, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25482198

RESUMO

At the onset mitosis in higher eukaryotes, the nuclear envelope (NE) undergoes dramatic deconstruction to allow separation of duplicated chromosomes. Studies have shown that during this process of nuclear envelope breakdown (NEBD), the extensive protein networks of the nuclear lamina are disassembled through phosphorylation of lamins and several inner nuclear membrane (INM) proteins. The LINC complex, composed of SUN and nesprin proteins, is involved in multiple interactions at the NE and plays vital roles in nuclear and cellular mechanics by connecting the nucleus to the cytoskeleton. Here, we show that SUN1, located in the INM, undergoes mitosis-specific phosphorylation on at least 3 sites within its nucleoplasmic N-terminus. We further identify Cdk1 as the kinase responsible for serine 48 and 333 phosphorylation, while serine 138 is phosphorylated by Plk1. In mitotic cells, SUN1 loses its interaction with N-terminal domain binding partners lamin A/C, emerin, and short nesprin-2 isoforms. Furthermore, a triple phosphomimetic SUN1 mutant displays increased solubility and reduced retention at the NE. In contrast, the central LINC complex interaction between the SUN1 C-terminus and the KASH domain of nesprin-2 is maintained during mitosis. Together, these data support a model whereby mitotic phosphorylation of SUN1 disrupts interactions with nucleoplasmic binding partners, promoting disassembly of the nuclear lamina and, potentially, its chromatin interactions. At the same time, our data add to an emerging picture that the core LINC complex plays an active role in NEBD.


Assuntos
Lamina Tipo A/metabolismo , Proteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/genética , Mitose/genética , Membrana Nuclear/genética , Proteínas Nucleares/genética , Proteína Quinase CDC2/metabolismo , Núcleo Celular/genética , Cromatina/genética , Células HeLa , Humanos , Lamina Tipo A/genética , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Lâmina Nuclear/genética , Proteínas Nucleares/metabolismo , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA