Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Aging (Albany NY) ; 12(20): 20658-20683, 2020 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-33098638

RESUMEN

Hormone receptor-positive breast cancer accounts for around 75% of breast cancers. The estrogen receptor pathway promotes tumor progression and endocrine resistance. Recently, the cross-talk between the ER signaling pathway and cell cycle regulation has been identified. It is necessary to determine the underlying molecular mechanisms involved in the ER signaling pathway and find new target genes for prognosis and drug resistance in ER+ breast cancer. In this study, lncRNA MAFG-AS1 was shown to be up-regulated and associated with poor prognosis in ER+ breast cancer. Functionally, down-regulation of MAFG-AS1 could inhibit cell proliferation and promote apoptosis. In addition, MAFG-AS1 which contained an estrogen-responsive element could promote CDK2 expression by sponging miR-339-5p. Subsequently, MAFG-AS1 and CDK2 were found to be up-regulated in tamoxifen-resistant MCF-7 cells. Cross-talk between the ER signaling pathway and cell cycle conducted by MAFG-AS1 and CDK2 could promote tamoxifen resistance. In conclusion, our study indicated that estrogen-responsive lncRNA MAFG-AS1 up-regulated CDK2 by sponging miR-339-5p, which promoted ER+ breast cancer proliferation. Cross-talk between the ER signaling pathway and cell cycle suggested that lncRNA MAFG-AS1 is a potential biomarker and therapeutic target in ER+ breast cancer. CDK2 inhibitors may be applied to endocrine resistance therapy.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Quinasa 2 Dependiente de la Ciclina/fisiología , Resistencia a Antineoplásicos , Factor de Transcripción MafG/fisiología , MicroARNs/fisiología , ARN Largo no Codificante , Receptor Cross-Talk , Receptores de Estrógenos/fisiología , Proteínas Represoras/fisiología , Transducción de Señal , Tamoxifeno/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Quinasa 2 Dependiente de la Ciclina/genética , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Factor de Transcripción MafG/genética , MicroARNs/genética , Persona de Mediana Edad , Proteínas Represoras/genética
2.
Biochem Biophys Res Commun ; 532(3): 400-405, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-32878707

RESUMEN

Hypoxia could cause vascular smooth muscle hypertrophy, leading to high pulmonary circulation resistance, pulmonary artery (PA) pressure, even pulmonary arterial hypertension (PAH). Recent studies have demonstrated the ability of mesenchymal stem cell (MSC) to ameliorate PAH but the mechanism was controversial. In this study, we revealed that the growth rate of pulmonary artery smooth muscle cells (PASMCs) treated with hypoxia was significantly increased than normal and showed lower expression of potassium channels. However, cells co-cultured with MSC showed decreased proliferation capability and down-regulated expression of ion channel of PAMSCs. The protein array data showed that the changes of PAMSCs was substantially associated with a high level of tumor necrosis factor alpha (TNFα) secretion from MSC. We further demonstrated that TNFα rescued the cell behavior of PAMSCs through activating the expression of P53 and NF-kB and inducing cell cycle arrest by P21/CDK2/CDK4 downregulation. These findings suggested that MSCs could attenuate abnormal function of PAMSCs by TNFα secretion, which was more or less associated with the beneficial effects of MSC on improving PAH.


Asunto(s)
Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/fisiopatología , Hipoxia/complicaciones , Hipoxia/fisiopatología , Células Madre Mesenquimatosas/fisiología , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Puntos de Control del Ciclo Celular , Técnicas de Cocultivo , Quinasa 2 Dependiente de la Ciclina/fisiología , Quinasa 4 Dependiente de la Ciclina/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Humanos , Hipertensión Pulmonar/patología , Hipoxia/patología , Células Madre Mesenquimatosas/patología , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Proteómica , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Transducción de Señal
3.
Transplantation ; 103(4): 724-732, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30801519

RESUMEN

BACKGROUND: Liver ischemia and reperfusion injury (IRI) is a major complication of liver transplant, hepatectomy, and hemorrhagic shock. The cyclin-dependent kinase 2 (CDK2) acts as a pivotal regulator of cell cycle and proliferation. METHODS: This study evaluated the modulation and therapeutic potential of CDK2 inhibition in a mouse model of partial liver warm IRI. RESULTS: Liver IR-triggered intrinsic CDK2 expression, peaking by 0.5 hour of reperfusion and maintaining a high-level throughout 1 to 24 hours. Roscovitine, a specific CDK2 inhibitor, prevented liver IR-mediated damage with abolished serum alanine aminotransferase levels and reserved liver pathology. CDK2 inhibition-mediated liver protection was accompanied by decreased macrophage/neutrophil infiltration, diminished hepatocyte apoptosis, abolished toll like receptor 4 signaling and downstream gene inductions (C-X-C motif ligand-10, Tumor necrosis factor-α, interleukin-1ß, and interleukin-6), yet augmented interleukin-10 expression. In vitro, CDK2 inhibition by Roscovitine suppressed macrophage TLR4 activation and further depressed downstream inflammatory signaling (myeloid differentiation factor 88, interferon regulatory transcription factor 3, p38, c-Jun N-terminal kinase, and extracellular-regulated kinase). CONCLUSIONS: Our novel findings revealed the critical role of CDK2 in hepatic cytoprotection and homeostasis against liver IRI. As CDK2 inhibition regulated local immune response and prevented hepatocyte death, this study provided the evidence for new treatment approaches to combat IRI in liver transplant.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Hígado/irrigación sanguínea , Daño por Reperfusión/prevención & control , Animales , Apoptosis , Quinasa 2 Dependiente de la Ciclina/fisiología , Sistema de Señalización de MAP Quinasas , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Transducción de Señal/fisiología , Receptor Toll-Like 4/fisiología
4.
Cell Syst ; 7(1): 17-27.e3, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-29909278

RESUMEN

Faithful DNA replication is challenged by stalling of replication forks during S phase. Replication stress is further increased in cancer cells or in response to genotoxic insults. Using live single-cell image analysis, we found that CDK2 activity fluctuates throughout an unperturbed S phase. We show that CDK2 fluctuations result from transient ATR signals triggered by stochastic replication stress events. In turn, fluctuating endogenous CDK2 activity causes corresponding decreases and increases in DNA synthesis rates, linking changes in stochastic replication stress to fluctuating global DNA replication rates throughout S phase. Moreover, cells that re-enter the cell cycle after mitogen stimulation have increased CDK2 fluctuations and prolonged S phase resulting from increased replication stress-induced CDK2 suppression. Thus, our study reveals a dynamic control principle for DNA replication whereby CDK2 activity is suppressed and fluctuates throughout S phase to continually adjust global DNA synthesis rates in response to recurring stochastic replication stress events.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , ADN/biosíntesis , Proteínas de la Ataxia Telangiectasia Mutada/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , División Celular , Línea Celular , Quinasa 2 Dependiente de la Ciclina/fisiología , Quinasas Ciclina-Dependientes/genética , Daño del ADN , Replicación del ADN , Proteínas de Unión al ADN/genética , Humanos , Células MCF-7 , Fase S/fisiología , Análisis de la Célula Individual/métodos
5.
Mol Cell ; 69(2): 253-264.e5, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29351845

RESUMEN

At the restriction point (R), mammalian cells irreversibly commit to divide. R has been viewed as a point in G1 that is passed when growth factor signaling initiates a positive feedback loop of Cdk activity. However, recent studies have cast doubt on this model by claiming R occurs prior to positive feedback activation in G1 or even before completion of the previous cell cycle. Here we reconcile these results and show that whereas many commonly used cell lines do not exhibit a G1 R, primary fibroblasts have a G1 R that is defined by a precise Cdk activity threshold and the activation of cell-cycle-dependent transcription. A simple threshold model, based solely on Cdk activity, predicted with more than 95% accuracy whether individual cells had passed R. That a single measurement accurately predicted cell fate shows that the state of complex regulatory networks can be assessed using a few critical protein activities.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/fisiología , Animales , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , División Celular , Línea Celular , Quinasa 2 Dependiente de la Ciclina/fisiología , Quinasas Ciclina-Dependientes/metabolismo , Fibroblastos/fisiología , Fase G1/fisiología , Humanos , Fosforilación , Cultivo Primario de Células , Transducción de Señal
6.
Nat Rev Cancer ; 17(2): 93-115, 2017 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-28127048

RESUMEN

Cancer is characterized by uncontrolled tumour cell proliferation resulting from aberrant activity of various cell cycle proteins. Therefore, cell cycle regulators are considered attractive targets in cancer therapy. Intriguingly, animal models demonstrate that some of these proteins are not essential for proliferation of non-transformed cells and development of most tissues. By contrast, many cancers are uniquely dependent on these proteins and hence are selectively sensitive to their inhibition. After decades of research on the physiological functions of cell cycle proteins and their relevance for cancer, this knowledge recently translated into the first approved cancer therapeutic targeting of a direct regulator of the cell cycle. In this Review, we focus on proteins that directly regulate cell cycle progression (such as cyclin-dependent kinases (CDKs)), as well as checkpoint kinases, Aurora kinases and Polo-like kinases (PLKs). We discuss the role of cell cycle proteins in cancer, the rationale for targeting them in cancer treatment and results of clinical trials, as well as the future therapeutic potential of various cell cycle inhibitors.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Animales , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/fisiología , Proteínas de Ciclo Celular/fisiología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/fisiología , Ensayos Clínicos como Asunto , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/fisiología , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/fisiología , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/fisiología , Humanos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/fisiología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal , Quinasa Tipo Polo 1
7.
Br J Cancer ; 115(6): 682-90, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27529512

RESUMEN

BACKGROUND: The phosphatidylinositol-3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway is commonly deregulated in human cancer, hence many PI3K and mTOR inhibitors have been developed and have now reached clinical trials. Similarly, CDKs have been investigated as cancer drug targets. METHODS: We have synthesised and characterised a series of 6-aminopyrimidines identified from a kinase screen that inhibit PI3K and/or mTOR and/or CDK2. Kinase inhibition, tumour cell growth, cell cycle distribution, cytotoxicity and signalling experiments were undertaken in HCT116 and HT29 colorectal cancer cell lines, and in vivo HT29 efficacy studies. RESULTS: 2,6-Diaminopyrimidines with an O(4)-cyclohexylmethyl substituent and a C-5-nitroso or cyano group (1,2,5) induced cell cycle phase alterations and were growth inhibitory (GI50<20 µM). Compound 1, but not 2 or 5, potently inhibits CDK2 (IC50=0.1 nM) as well as PI3K, and was cytotoxic at growth inhibitory concentrations. Consistent with kinase inhibition data, compound 1 reduced phospho-Rb and phospho-rS6 at GI50 concentrations. Combination of NU6102 (CDK2 inhibitor) and pictilisib (GDC-0941; pan-PI3K inhibitor) resulted in synergistic growth inhibition, and enhanced cytotoxicity in HT29 cells in vitro and HT29 tumour growth inhibition in vivo. CONCLUSIONS: These studies identified a novel series of mixed CDK2/PI3K inhibitors and demonstrate that dual targeting of CDK2 and PI3K can result in enhanced antitumour activity.


Asunto(s)
Adenocarcinoma/patología , Neoplasias Colorrectales/patología , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Adenocarcinoma/enzimología , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Colorrectales/enzimología , Quinasa 2 Dependiente de la Ciclina/fisiología , Humanos , Ratones , Ratones Desnudos , Proteínas de Neoplasias/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/síntesis química , Pirimidinas/farmacología , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Carcinogenesis ; 37(9): 858-869, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27496804

RESUMEN

miR-200b is a pleiotropically acting microRNA in cancer progression, representing an attractive therapeutic target. We previously identified miR-200b as an invasiveness repressor in esophageal squamous cell carcinoma (ESCC), whereas further understanding is warranted to establish it as a therapeutic target. Here, we show that miR-200b mitigates ESCC cell growth by inducing G2-phase cell cycle arrest and apoptosis. The expression/activation of multiple key cell cycle regulators such as CDK1, CDK2, CDK4 and Cyclin B, and the Wnt/ß-Catenin signaling are modulated by miR-200b. We identified CDK2 and PAF (PCNA-associated factor), two important tumor-promoting factors, as direct miR-200b targets in ESCC. Correlating with the frequent loss of miR-200b in ESCC, both CDK2 and PAF levels are significantly increased in ESCC tumors compared to case-matched normal tissues (n = 119, both P < 0.0001), and correlate with markedly reduced survival (P = 0.007 and P = 0.041, respectively). Furthermore, CDK2 and PAF are also associated with poor prognosis in certain subtypes of breast cancer (n = 1802) and gastric cancer (n = 233). Although CDK2 could not significantly mediate the biological function of miR-200b, PAF siRNA knockdown phenocopied while restored expression of PAF abrogated the biological effects of miR-200b on ESCC cells. Moreover, PAF was revealed to mediate the inhibitory effects of miR-200b on Wnt/ß-Catenin signaling. Collectively, the pleiotropic effects of miR-200b in ESCC highlight its potential for therapeutic intervention in this aggressive disease.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/patología , Puntos de Control de la Fase G2 del Ciclo Celular , MicroARNs/fisiología , Apoptosis , Carcinoma de Células Escamosas/genética , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Línea Celular Tumoral , Proliferación Celular , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/fisiología , Proteínas de Unión al ADN , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago , Genes Supresores de Tumor , Humanos , Pronóstico , Vía de Señalización Wnt , beta Catenina/fisiología
9.
Gynecol Oncol ; 143(1): 152-158, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27461360

RESUMEN

Cyclin E1 (CCNE1) gene amplification occurs in approximately 20% of ovarian high grade serous carcinoma (HGSC) and is associated with chemotherapy resistance and, in some studies, overall poor prognosis. The role of cyclin E1 in inducing S phase entry relies upon its interactions with cyclin dependent kinases (CDK), specifically CDK2. Therapies to target cyclin E1-related functions have centered on CDK inhibitors and proteasome inhibitors. While many studies have helped elucidate the functions and regulatory mechanisms of cyclin E1, further research utilizing cyclin E1 as a therapeutic target in ovarian cancer is warranted. This review serves to present the scientific background describing the role and function of cyclin E1 in cancer development and progression, to distinguish cyclin E1-amplified HGSC as a unique subset of ovarian cancer deserving of further therapeutic investigation, and to provide an updated overview on the studies which have utilized cyclin E1 as a target for therapy in ovarian cancer.


Asunto(s)
Ciclina E/fisiología , Cistadenocarcinoma Seroso/etiología , Proteínas Oncogénicas/fisiología , Neoplasias Ováricas/etiología , Ciclina E/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/fisiología , Cistadenocarcinoma Seroso/terapia , Femenino , Humanos , Proteínas Oncogénicas/antagonistas & inhibidores , Neoplasias Ováricas/terapia
10.
Tumour Biol ; 37(9): 12485-12495, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27337954

RESUMEN

Increased expression of insulin-like growth factor 2 (IGF2) is found in tumors of colorectal cancer (CRC) patients exhibiting a gained region on chromosome 11q15 and is implicated in poor patient survival. This study analyzes in vitro phenotypic- and gene expression changes associated with IGF2 shRNA-mediated knockdown. Initially, doxycycline inducible IGF2 knockdown cell lines were generated in the CRC cell lines SW480 and LS174T. The cells were analyzed for changes in proliferation, cell cycle, apoptosis, adhesion, and invasion. Expression profiling analysis was performed, and, for a subset of the identified genes, expression was validated by qRT-PCR and Western blot. IGF2 knockdown inhibited cell proliferation in both cell lines induced G1 cell cycle blockade and decreased adhesion to several extracellular matrix proteins. Knockdown of IGF2 did not alter invasiveness in SW480 cells, while a slight increase in apoptosis was seen only in the LS174T cell line. Knockdown of IGF2 in SW480 deregulated 58 genes, several of which were associated with proliferation and cell-cell/cell-ECM contacts. A subset of these genes, including CDK2, YAP1, and BIRC5 (Survivin), are members of a common network. This study supports the concept of direct autocrine/paracrine tumor cell activation through IGF2 and a shows role of IGF2 in CRC proliferation, adhesion and, to a limited extent, apoptosis.


Asunto(s)
Neoplasias Colorrectales/patología , Factor II del Crecimiento Similar a la Insulina/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Antígenos CD , Cadherinas/genética , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Quinasa 2 Dependiente de la Ciclina/fisiología , Desmoplaquinas/genética , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Fosfoproteínas/genética , ARN Interferente Pequeño/genética , Factores de Transcripción , Proteínas Señalizadoras YAP , gamma Catenina
11.
J Proteomics ; 141: 77-84, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27109354

RESUMEN

UNLABELLED: The epidermal growth factor (EGF) receptor (EGFR) pathway is one of the most dysregulated and extensively investigated signaling pathways in human cancers and plays important roles in the regulation of nuclear functions through both cytoplasmic and nuclear EGFR pathways. However, the current understanding of the nuclear phosphorylation responses to activated EGFR pathways remains limited. In the present study, phosphoproteomics analysis revealed the increased phosphorylation of 90 nuclear proteins, primarily involved in RNA processing, pre-mRNA splicing and cell cycle regulation, upon EGF stimulation in MDA-MB-468 cells. Cellular splicing assays of the ß-globin (HBB) minigene confirmed that EGF induced constitutive pre-mRNA splicing. Further analysis of phosphoproteomics data identified multiple CDK1/2 substrates in pre-mRNA splicing-related proteins, and both CDK1/2 inhibitors and CDK1/2 knockdowns reduced EGF-regulated pre-mRNA splicing. In conclusion, the results of the present study provide evidence that CDK1/2 participate in the regulation of constitutive pre-mRNA splicing by EGF stimulation in MDA-MB-468 cells. SIGNIFICANCE: In this study, we successfully carried out a survey of nuclear phosphorylation changes in response to EGF stimulation. The results from the functional category analysis and pre-mRNA splicing assay strongly indicated that EGFR activation increased constitutive pre-mRNA splicing in MDA-MB-468 cells, revealing additional role of EGFR on regulation of mRNA maturation beyond alternative pre-mRNA splicing reported by previous studies. Furthermore, we found that CDK1/2 participated in constitutive pre-mRNA splicing regulation by EGF in MDA-MB-468 cells. Our study provides new knowledge for understanding the regulation of constitutive pre-mRNA splicing by EGF stimulation.


Asunto(s)
Proteína Quinasa CDC2/fisiología , Quinasa 2 Dependiente de la Ciclina/fisiología , Factor de Crecimiento Epidérmico/fisiología , Proteínas Nucleares/metabolismo , Empalme del ARN , Línea Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Fosforilación/efectos de los fármacos , Precursores del ARN
12.
Cell Cycle ; 15(1): 137-51, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26771717

RESUMEN

Expression of Breast Cancer Metastasis Suppressor 1 (BRMS1) reduces the incidence of metastasis in many human cancers, without affecting tumorigenesis. BRMS1 carries out this function through several mechanisms, including regulation of gene expression by binding to the mSin3/histone deacetylase (HDAC) transcriptional repressor complex. In the present study, we show that BRMS1 is a novel substrate of Cyclin-Dependent Kinase 2 (CDK2) that is phosphorylated on serine 237 (S237). Although CDKs are known to regulate cell cycle progression, the mutation of BRMS1 on serine 237 did not affect cell cycle progression and proliferation of MDA-MB-231 breast cancer cells; however, their migration was affected. Phosphorylation of BRMS1 does not affect its association with the mSin3/HDAC transcriptional repressor complex or its transcriptional repressor activity. The serine 237 phosphorylation site is immediately proximal to a C-terminal nuclear localization sequence that plays an important role in BRMS1-mediated metastasis suppression but phosphorylation does not control BRMS1 subcellular localization. Our studies demonstrate that CDK-mediated phosphorylation of BRMS1 regulates the migration of tumor cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular/fisiología , Quinasa 2 Dependiente de la Ciclina/fisiología , Proteínas Represoras/metabolismo , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Fosforilación/fisiología
13.
Tumour Biol ; 35(4): 3339-46, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24307622

RESUMEN

This study aimed to analyze the expression, clinical significance of cyclin G2 (CCNG2) in colorectal carcinoma, and the biological effect in its cell line by CCNG2 overexpression. Immunohistochemistry and Western blot were used to analyze CCNG2 protein expression in colorectal cancer and to study the influence of the upregulated expression of CCNG2 that might be found on SW480 cell biological effect. We found that the level of CCNG2 protein expression was significantly lower in colorectal cancer tissue than normal tissues (P < 0.05). The level of CCNG2 was correlated with T stages, lymph node metastasis, clinic stage, and histological grade (P < 0.05). Loss of CCNG2 expression correlated significantly with poor overall survival time by Kaplan-Meier analysis (P < 0.05). The result of biological function has shown that SW480 cell-transfected CCNG2 had a lower survival fraction, higher percentage of the G0/G1 phases, and lower CDK2 protein expression compared with SW480 cell-untransfected CCNG2 (P < 0.05).


Asunto(s)
Neoplasias Colorrectales/patología , Ciclina G2/fisiología , Adulto , Anciano , Ciclo Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/mortalidad , Ciclina G2/análisis , Ciclina G2/genética , Quinasa 2 Dependiente de la Ciclina/fisiología , Regulación hacia Abajo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico
14.
Hepatology ; 59(2): 651-60, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23787781

RESUMEN

UNLABELLED: The liver has a strong regenerative capacity. After injury, quiescent hepatocytes can reenter the mitotic cell cycle to restore tissue homeostasis. This G(0) /G(1) -S cell-cycle transition of primed hepatocytes is regulated by complexes of cyclin-dependent kinase 2 (Cdk2) with E-type cyclins (CcnE1 or CcnE2). However, single genetic ablation of either E-cyclin or Cdk2 does not affect overall liver regeneration. Here, we systematically investigated the contribution of CcnE1, CcnE2, and Cdk2 for liver regeneration after partial hepatectomy (PH) by generating corresponding double- and triple-knockout (KO) mouse mutants. We demonstrate that conditional deletion of Cdk2 alone in hepatocytes resulted in accelerated induction of CcnE1, but otherwise normal initiation of S phase in vivo and in vitro. Excessive CcnE1 did not contribute to a noncanonical kinase activity, but was located at chromatin together with components of the pre-replication complex (pre-RC), such as the minichromosome maintenance (MCM) helicase. Concomitant ablation of Cdk2 and CcnE1 in hepatocytes caused a defect in pre-RC formation and further led to dramatically impaired S-phase progression by down-regulation of cyclin A2 and cell death in vitro and substantially reduced hepatocyte proliferation and liver regeneration after PH in vivo. Similarly, combined loss of CcnE1 and CcnE2, but also the Cdk2/CcnE1/CcnE2 triple KO in liver, significantly inhibited S-phase initiation and liver mass reconstitution after PH, whereas concomitant ablation of CcnE2 and Cdk2 had no effect. CONCLUSION: In the absence of Cdk2, CcnE1 performs crucial kinase-independent functions in hepatocytes, which are capable of driving MCM loading on chromatin, cyclin A2 expression, and S-phase progression. Thus, combined inactivation of Cdk2 and CcnE1 is the minimal requirement for blocking S-phase machinery in vivo.


Asunto(s)
Ciclina E/deficiencia , Quinasa 2 Dependiente de la Ciclina/deficiencia , Replicación del ADN/fisiología , Hepatocitos/patología , Hepatocitos/fisiología , Regeneración Hepática/fisiología , Proteínas Oncogénicas/deficiencia , Animales , Apoptosis/fisiología , Ciclo Celular/fisiología , Células Cultivadas , Cromatina/fisiología , Ciclina E/genética , Ciclina E/fisiología , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/fisiología , Ciclinas/deficiencia , Ciclinas/genética , Ciclinas/fisiología , Femenino , Homeostasis/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/fisiología , Fase S/fisiología
15.
Cold Spring Harb Perspect Biol ; 5(1): a012948, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23284048

RESUMEN

Developmentally programmed polyploidy occurs by at least four different mechanisms, two of which (endoreduplication and endomitosis) involve switching from mitotic cell cycles to endocycles by the selective loss of mitotic cyclin-dependent kinase (CDK) activity and bypassing many of the processes of mitosis. Here we review the mechanisms of endoreplication, focusing on recent results from Drosophila and mice.


Asunto(s)
Drosophila/genética , Endorreduplicación/fisiología , Animales , Ciclo Celular/fisiología , Ciclina E/metabolismo , Ciclina E/fisiología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 2 Dependiente de la Ciclina/fisiología , Replicación del ADN , Drosophila/citología , Ratones , Poliploidía
16.
Int J Biol Sci ; 8(10): 1431-46, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23197940

RESUMEN

Carbon monoxide (CO) is a vasoactive molecule that is generated by vascular cells as a byproduct of heme catabolism and it plays an important physiological role in circulation system. In order to investigate whether exogenous CO can mediate the growth and proliferation of vascular cells, in this study, we used 250 parts per million (ppm) of CO to treat human umbilical artery smooth muscle cell (hUASMC) and human umbilical vein endothelial cell (HuVEC) and further evaluated the growth and apoptosis status of SMC and HuVEC. After SMC and HuVEC were exposed to CO for 7-day, the growth of SMC and HuVEC was significantly inhibited by CO in vitro on day 5 of CO exposure. And CO blocked cell cycle progress of SMC and HuVEC, more SMC and HuVEC stagnated at G0/G1 phase by flow cytometric analysis. Moreover, CO treatment inhibited SMC and HuVEC apoptosis caused by hydrogen peroxide through decreasing caspase 3 and 9 activities. To confirm the molecular mechanism of CO effect on SMC and HuVEC growth, we compared the gene expression profile in SMC and CO-treated SMC, HuVEC and CO-treated HuVEC. By microarray analysis, we found the expression level of some genes which are related to cell cycle regulation, cell growth and proliferation, and apoptosis were changed during CO exposure. We further identified that the down-regulated CDK2 contributed to arresting cell growth and the down-regulated Caspase 3 (CASP3) and Caspase 9 (CASP9) were associated with the inhibition of cell apoptosis. Therefore, CO exerts a certain growth arrest on SMC and HuVEC by inhibiting cell cycle transition from G0/G1 phase to S phase and has regulatory effect on cell apoptosis by regulating the expression of apoptosis-associated genes.


Asunto(s)
Apoptosis/efectos de los fármacos , Monóxido de Carbono/farmacología , Células Endoteliales/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Arterias Umbilicales/efectos de los fármacos , Apoptosis/genética , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Transducción de Señal , Arterias Umbilicales/metabolismo
17.
J Immunol ; 189(12): 5659-66, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23136201

RESUMEN

Adaptive immunity requires signals from both the TCR and the costimulatory molecule CD28. These receptors activate multiple signaling pathways, including the cyclin-dependent kinase (CDK) cascade, and antigenic signals in the absence of costimulation result in a tolerant state that is enforced by the CDK inhibitory protein p27kip1. We find that CDK2, the major target of p27kip1, is highly active in T cells that infiltrate and reject cardiac allografts. We used mice genetically deficient for CDK2 to determine whether CDK2 is required for T cell alloimmunity. Blockade of CD28 costimulation alone was unable to inhibit the rejection of cardiac allografts by wild-type recipients. However, targeting this pathway in CDK2-deficient recipients led to long-term allograft survival. CDK2-deficient CD4(+) T cells proliferated normally in response to stimulation in vitro and in vivo, however, genetic, short hairpin RNA, or small molecule-mediated antagonism of CDK2 resulted in decreased production of IL-2 and IFN-γ. In addition, surviving grafts from CDK2-deficient recipients showed increased infiltration of Foxp3(+) regulatory T cells (Treg), and Treg from CDK2-deficient mice exhibited increased suppressive activity in vitro and in an in vivo model of inflammatory bowel disease. These data suggest that p27kip1 promotes peripheral tolerance through its ability to inhibit CDK2, which otherwise acts to promote conventional T cell differentiation and restrict Treg function.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/fisiología , Animales , Células Cultivadas , Colitis/enzimología , Colitis/inmunología , Colitis/patología , Quinasa 2 Dependiente de la Ciclina/deficiencia , Quinasa 2 Dependiente de la Ciclina/genética , Modelos Animales de Enfermedad , Femenino , Tolerancia Inmunológica/genética , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
18.
Proc Natl Acad Sci U S A ; 109(17): E1019-27, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22474407

RESUMEN

A family of conserved serine/threonine kinases known as cyclin-dependent kinases (CDKs) drives orderly cell cycle progression in mammalian cells. Prior studies have suggested that CDK2 regulates S-phase entry and progression, and frequently shows increased activity in a wide spectrum of human tumors. Genetic KO/knockdown approaches, however, have suggested that lack of CDK2 protein does not prevent cellular proliferation, both during somatic development in mice as well as in human cancer cell lines. Here, we use an alternative, chemical-genetic approach to achieve specific inhibition of CDK2 kinase activity in cells. We directly compare small-molecule inhibition of CDK2 kinase activity with siRNA knockdown and show that small-molecule inhibition results in marked defects in proliferation of nontransformed cells, whereas siRNA knockdown does not, highlighting the differences between these two approaches. In addition, CDK2 inhibition drastically diminishes anchorage-independent growth of human cancer cells and cells transformed with various oncogenes. Our results establish that CDK2 activity is necessary for normal mammalian cell cycle progression and suggest that it might be a useful therapeutic target for treating cancer.


Asunto(s)
Transformación Celular Neoplásica , Quinasa 2 Dependiente de la Ciclina/fisiología , Oncogenes , Animales , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/química , Quinasa 2 Dependiente de la Ciclina/genética , Técnicas de Silenciamiento del Gen , Humanos , Ratones , ARN Interferente Pequeño
19.
Oncogene ; 31(48): 5019-28, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-22286767

RESUMEN

RB(+/-) individuals develop retinoblastoma and, subsequently, many other tumors. The Rb relatives p107 and p130 protect the tumor-resistant Rb(-/-) mouse retina. Determining the mechanism underlying this tumor suppressor function may expose novel strategies to block Rb pathway cancers. p107/p130 are best known as E2f inhibitors, but here we implicate E2f-independent Cdk2 inhibition as the critical p107 tumor suppressor function in vivo. Like p107 loss, deleting p27 or inactivating its Cdk inhibitor (CKI) function (p27(CK-)) cooperated with Rb loss to induce retinoblastoma. Genetically, p107 behaved like a CKI because inactivating Rb and one allele each of p27 and p107 was tumorigenic. Although Rb loss induced canonical E2f targets, unexpectedly p107 loss did not further induce these genes, but instead caused post-transcriptional Skp2 induction and Cdk2 activation. Strikingly, Cdk2 activity correlated with tumor penetrance across all the retinoblastoma models. Therefore, Rb restrains E2f, but p107 inhibits cross talk to Cdk. While removing either E2f2 or E2f3 genes had little effect, removing only one E2f1 allele blocked tumorigenesis. More importantly, exposing retinoblastoma-prone fetuses to small molecule inhibitors of E2f (HLM006474) or Cdk (R547) for merely 1 week dramatically inhibited subsequent tumorigenesis in adult mice. Protection was achieved without disrupting normal proliferation. Thus, exquisite sensitivity of the cell-of-origin to E2f and Cdk activity can be exploited to prevent Rb pathway-induced cancer in vivo without perturbing normal cell division. These data suggest that E2f inhibitors, never before tested in vivo, or CKIs, largely disappointing as therapeutics, may be effective preventive agents.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/fisiología , Factor de Transcripción E2F1/fisiología , Retinoblastoma/fisiopatología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Retinoblastoma/patología , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Mol Cell ; 42(5): 624-36, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21658603

RESUMEN

Multiple cyclin-dependent kinases (CDKs) control eukaryotic cell division, but assigning specific functions to individual CDKs remains a challenge. During the mammalian cell cycle, Cdk2 forms active complexes before Cdk1, but lack of Cdk2 protein does not block cell-cycle progression. To detect requirements and define functions for Cdk2 activity in human cells when normal expression levels are preserved, and nonphysiologic compensation by other CDKs is prevented, we replaced the wild-type kinase with a version sensitized to specific inhibition by bulky adenine analogs. The sensitizing mutation also impaired a noncatalytic function of Cdk2 in restricting assembly of cyclin A with Cdk1, but this defect could be corrected by both inhibitory and noninhibitory analogs. This allowed either chemical rescue or selective antagonism of Cdk2 activity in vivo, to uncover a requirement in cell proliferation, and nonredundant, rate-limiting roles in restriction point passage and S phase entry.


Asunto(s)
Proliferación Celular , Quinasa 2 Dependiente de la Ciclina/fisiología , Adenina/análogos & derivados , Adenina/farmacología , Línea Celular , Quinasa 2 Dependiente de la Ciclina/química , Quinasa 2 Dependiente de la Ciclina/genética , Fase G1/efectos de los fármacos , Fase G1/fisiología , Humanos , Estructura Terciaria de Proteína , Fase S/efectos de los fármacos , Fase S/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA