Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Exp Neurol ; 340: 113686, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33713658

RESUMO

BACKGROUND AND PURPOSE: Mast cells (MCs) has been recognized as an effector of inflammation or a trigger of inflammatory factors during stroke. LJ529 was reported to attenuate inflammation through a Gi protein-coupled Adenosine A3 receptor (A3R) after ischemia. Here, we aim to study the protective effect and its mechanism of LJ529 in subarachnoid hemorrhage (SAH) rat model for mast cell-related inflammation. METHODS: 155 Sprague-Dawley adult male rats were used in experiments. Endovascular perforation was used for SAH model. Intraperitoneal LJ529 was performed 1 h after SAH. Neurological scores were measured 24 h after SAH. Rotarod and morris water maze tests were evaluated for 21 days after SAH. Mast cell degranulation was assessed with Toluidine blue staining and Chymase/Typtase protein expressions. Mast cell-related inflammation was evaluated using IL-6, TNF-α and MCP-1 protein expressions. MRS1523, inhibitor of GPR18 and ε-V1-2, inhibitor of PKCε were respectively given intraperitoneally (i.p.) 1 h and 30 min before SAH for mechanism studies. Pathway related proteins were investigated with western blot and immunofluorescence staining. RESULTS: Expression of A3R, PKCε increased after SAH. LJ529 treatment attenuated mast cell degranulation and inflammation. Meanwhile, both short-term and long-term neurological functions were improved after LJ529 treatment. Administration of LJ529 resulted in increased expressions of A3R, PKCε, ALDH2 proteins and decreased expressions of Chymase, Typtase, IL-6, TNF-α and MCP-1 proteins. MRS1523 abolished the treatment effects of LJ529 on neurobehavior and protein levels. ε-V1-2 also reversed the outcomes of LJ529 administration through reduction in protein expressions downstream of PKCε. CONCLUSIONS: LJ529 attenuated mast cell-related inflammation through inhibiting degranulation via A3R-PKCε-ALDH2 pathway after SAH. LJ529 may serve as a potential treatment strategy to relieve post-SAH brain injury.


Assuntos
Agonistas do Receptor A3 de Adenosina/uso terapêutico , Adenosina/análogos & derivados , Aldeído-Desidrogenase Mitocondrial/biossíntese , Proteína Quinase C-épsilon/biossíntese , Receptor A3 de Adenosina/biossíntese , Hemorragia Subaracnóidea/tratamento farmacológico , Tionucleosídeos/uso terapêutico , Adenosina/farmacologia , Adenosina/uso terapêutico , Agonistas do Receptor A3 de Adenosina/farmacologia , Animais , Relação Dose-Resposta a Droga , Inflamação/metabolismo , Inflamação/prevenção & controle , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Hemorragia Subaracnóidea/metabolismo , Tionucleosídeos/farmacologia
2.
Cell Death Dis ; 8(5): e2770, 2017 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-28492560

RESUMO

Gallbladder cancer (GBC) is one of the most common malignancy of the biliary tract characterized by its high chemoresistant tendency. Although great progresses have been made in recent decades for treating many cancers with anticancer drugs, effective therapeutics methods for anti-GBC are still lacking. Therefore, investigations into identifying the mechanisms underlying the drug resistance of GBC are greatly needed. In this study, we show that miR-218-5p plays a critical role in gemcitabine resistance of GBC. miR-218-5p levels were significantly lower in GBC than adjacent non-cancer tissues, and which were also associated with patient prognosis. While miR-218-5p overexpression abrogated gemcitabine resistance of GBC cells, silencing of which exhibited the opposite effects. Via six microRNA targets prediction algorithms, we found that PRKCE is a potential target of miR-218-5p. Moreover, miR-218-5p overexpression repressed the luciferase activity of reporter constructs containing 3'-UTR of PRKCE and also reduced PRKCE expression. Further studies revealed that miR-218-5p promotes sensitivity of gemcitabine by abolishing PRKCE-induced upregulation of MDR1/P-gp. Taken together, our results imply that an intimate correlation between miR-218-5p and PRKCE/MDR1 axis abnormal expression is a key determinant of gemcitabine tolerance, and suggest a novel miR-218-5p-based clinical intervention target for GBC patients.


Assuntos
Desoxicitidina/análogos & derivados , Neoplasias da Vesícula Biliar , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , MicroRNAs , Proteínas de Neoplasias , Proteína Quinase C-épsilon , RNA Neoplásico , Regulação para Cima/efeitos dos fármacos , Subfamília B de Transportador de Cassetes de Ligação de ATP/biossíntese , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Desoxicitidina/farmacologia , Feminino , Neoplasias da Vesícula Biliar/tratamento farmacológico , Neoplasias da Vesícula Biliar/genética , Neoplasias da Vesícula Biliar/metabolismo , Neoplasias da Vesícula Biliar/patologia , Humanos , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteína Quinase C-épsilon/biossíntese , Proteína Quinase C-épsilon/genética , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Gencitabina
3.
Cell Rep ; 19(2): 375-388, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28402859

RESUMO

PKCε, an oncogenic member of the PKC family, is aberrantly overexpressed in epithelial cancers. To date, little is known about functional interactions of PKCε with other genetic alterations, as well as the effectors contributing to its tumorigenic and metastatic phenotype. Here, we demonstrate that PKCε cooperates with the loss of the tumor suppressor Pten for the development of prostate cancer in a mouse model. Mechanistic analysis revealed that PKCε overexpression and Pten loss individually and synergistically upregulate the production of the chemokine CXCL13, which involves the transcriptional activation of the CXCL13 gene via the non-canonical nuclear factor κB (NF-κB) pathway. Notably, targeted disruption of CXCL13 or its receptor, CXCR5, in prostate cancer cells impaired their migratory and tumorigenic properties. In addition to providing evidence for an autonomous vicious cycle driven by PKCε, our studies identified a compelling rationale for targeting the CXCL13-CXCR5 axis for prostate cancer treatment.


Assuntos
Quimiocina CXCL13/genética , PTEN Fosfo-Hidrolase/biossíntese , Neoplasias da Próstata/genética , Proteína Quinase C-épsilon/biossíntese , Receptores CXCR5/genética , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Movimento Celular , Quimiocina CXCL13/biossíntese , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , NF-kappa B , Metástase Neoplásica , PTEN Fosfo-Hidrolase/genética , Neoplasias da Próstata/patologia , Proteína Quinase C-épsilon/genética , Receptores CXCR5/biossíntese , Transdução de Sinais
4.
PLoS One ; 11(2): e0150557, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26918336

RESUMO

Fetal nicotine exposure increased risk of developing cardiovascular disease later in life. The present study tested the hypothesis that perinatal nicotine-induced programming of heart ischemia-sensitive phenotype is mediated by enhanced reactive oxygen species (ROS) in offspring. Nicotine was administered to pregnant rats via subcutaneous osmotic minipumps from day 4 of gestation to day 10 after birth, in the absence or presence of a ROS inhibitor, N-acetyl-cysteine (NAC) in drinking water. Experiments were conducted in 8 month old age male offspring. Isolated hearts were perfused in a Langendorff preparation. Perinatal nicotine treatment significantly increased ischemia and reperfusion-induced left ventricular injury, and decreased post-ischemic recovery of left ventricular function and coronary flow rate. In addition, nicotine enhanced cardiac ROS production and significantly attenuated protein kinase Cε (PKCε) protein abundance in the heart. Although nicotine had no effect on total cardiac glycogen synthase kinase-3ß (GSK3ß) protein expression, it significantly increased the phosphorylation of GSK3ß at serine 9 residue in the heart. NAC inhibited nicotine-mediated increase in ROS production, recovered PKCε gene expression and abrogated increased phosphorylation of GSK3ß. Of importance, NAC blocked perinatal nicotine-induced increase in ischemia and reperfusion injury in the heart. These findings provide novel evidence that increased oxidative stress plays a causal role in perinatal nicotine-induced developmental programming of ischemic sensitive phenotype in the heart, and suggest potential therapeutic targets of anti-oxidative stress in the treatment of ischemic heart disease.


Assuntos
Acetilcisteína/uso terapêutico , Antioxidantes/uso terapêutico , Isquemia Miocárdica/etiologia , Nicotina/toxicidade , Efeitos Tardios da Exposição Pré-Natal , Disfunção Ventricular Esquerda/etiologia , Acetilcisteína/administração & dosagem , Animais , Antioxidantes/administração & dosagem , Circulação Coronária/efeitos dos fármacos , Suscetibilidade a Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Feto/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Bombas de Infusão Implantáveis , Masculino , Modelos Biológicos , Isquemia Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/etiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Nicotina/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Fenótipo , Fosforilação/efeitos dos fármacos , Gravidez , Proteína Quinase C-épsilon/biossíntese , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio , Recuperação de Função Fisiológica , Disfunção Ventricular Esquerda/prevenção & controle
5.
Carcinogenesis ; 37(1): 72-80, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26586792

RESUMO

Protein kinase C epsilon (PKCε), a Ca(2+)-independent phospholipid-dependent serine/threonine kinase, is among the six PKC isoforms (α, δ, ε, η, µ, ζ) expressed in both mouse and human skin. Epidermal PKCε level dictates the susceptibility of PKCε transgenic (TG) mice to the development of cutaneous squamous cell carcinomas (SCC) elicited either by repeated exposure to ultraviolet radiation (UVR) or by using the DMBA initiation-TPA (12-O-tetradecanoylphorbol-13-acetate) tumor promotion protocol (Wheeler,D.L. et al. (2004) Protein kinase C epsilon is an endogenous photosensitizer that enhances ultraviolet radiation-induced cutaneous damage and development of squamous cell carcinomas. Cancer Res., 64, 7756-7765). Histologically, SCC in TG mice, like human SCC, is poorly differentiated and metastatic. Our earlier studies to elucidate mechanisms of PKCε-mediated development of SCC, using either DMBA-TPA or UVR, indicated elevated release of cytokine TNFα. To determine whether TNFα is essential for the development of SCC in TG mice, we generated PKCε transgenic mice/TNFα-knockout (TG/TNFαKO) by crossbreeding TNFαKO with TG mice. We now present that deletion of TNFα in TG mice inhibited the development of SCC either by repeated UVR exposures or by the DMBA-TPA protocol. TG mice deficient in TNFα elicited both increase in SCC latency and decrease in SCC incidence. Inhibition of UVR-induced SCC development in TG/TNFαKO was accompanied by inhibition of (i) the expression levels of TNFα receptors TNFRI and TNFRII and cell proliferation marker ornithine decarboxylase and metastatic markers MMP7 and MMP9, (ii) the activation of transcription factors Stat3 and NF-kB and (iii) proliferation of hair follicle stem cells and epidermal hyperplasia. The results presented here provide the first genetic evidence that TNFα is linked to PKCε-mediated sensitivity to DMBA-TPA or UVR-induced development of cutaneous SCC.


Assuntos
Carcinoma de Células Escamosas/prevenção & controle , Proteína Quinase C-épsilon/genética , Neoplasias Cutâneas/prevenção & controle , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinogênese/induzido quimicamente , Carcinogênese/genética , Carcinogênese/efeitos da radiação , Carcinógenos , Carcinoma de Células Escamosas/etiologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Feminino , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteína Quinase C-épsilon/biossíntese , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Acetato de Tetradecanoilforbol , Raios Ultravioleta
6.
Am J Physiol Heart Circ Physiol ; 310(4): H516-23, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26683901

RESUMO

Methamphetamine is one of the most common illicit drugs abused during pregnancy. The neurological effects of prenatal methamphetamine are well known. However, few studies have investigated the potential effects of prenatal methamphetamine on adult cardiovascular function. Previous work demonstrated that prenatal cocaine exposure increases sensitivity of the adult heart to ischemic injury. Methamphetamine and cocaine have different mechanisms of action, but both drugs exert their effects by increasing dopaminergic and adrenergic receptor stimulation. Thus the goal of this study was to determine whether prenatal methamphetamine also worsens ischemic injury in the adult heart. Pregnant rats were injected with methamphetamine (5 mg·kg(-1)·day(-1)) or saline throughout pregnancy. When pups reached 8 wk of age, their hearts were subjected to ischemia and reperfusion by means of a Langendorff isolated heart system. Prenatal methamphetamine had no significant effect on infarct size, preischemic contractile function, or postischemic recovery of contractile function in male hearts. However, methamphetamine-treated female hearts exhibited significantly larger infarcts and significantly elevated end-diastolic pressure during recovery from ischemia. Methamphetamine significantly reduced protein kinase Cε expression and Akt phosphorylation in female hearts but had no effect on these cardioprotective proteins in male hearts. These data indicate that prenatal methamphetamine differentially affects male and female sensitivity to myocardial ischemic injury and alters cardioprotective signaling proteins in the adult heart.


Assuntos
Estimulantes do Sistema Nervoso Central/toxicidade , Metanfetamina/toxicidade , Isquemia Miocárdica/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Animais , Peso ao Nascer/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Feminino , Técnicas In Vitro , Masculino , Atividade Motora/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/induzido quimicamente , Infarto do Miocárdio/patologia , Isquemia Miocárdica/induzido quimicamente , Traumatismo por Reperfusão Miocárdica/patologia , Proteína Oncogênica v-akt/metabolismo , Fosforilação/efeitos dos fármacos , Gravidez , Proteína Quinase C-épsilon/biossíntese , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais
7.
Cell Death Dis ; 6: e1758, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25950488

RESUMO

RIG-I-like receptors are the key cytosolic sensors for RNA viruses and induce the production of type I interferons (IFN) and pro-inflammatory cytokines through a sole adaptor IFN-ß promoter stimulator-1 (IPS-1) (also known as Cardif, MAVS and VISA) in antiviral innate immunity. These sensors also have a pivotal role in anticancer activity through induction of apoptosis. However, the mechanism for their anticancer activity is poorly understood. Here, we show that anticancer vaccine adjuvant, PolyIC (primarily sensed by MDA5) and the oncolytic virus, Newcastle disease virus (NDV) (sensed by RIG-I), induce anticancer activity. The ectopic expression of IPS-1 into type I IFN-responsive and non-responsive cancer cells induces anticancer activity. PolyIC transfection and NDV infection upregulate pro-apoptotic gene TRAIL and downregulate the anti-apoptotic genes BCL2, BIRC3 and PRKCE. Furthermore, stable knockdown of IPS-1, IRF3 or IRF7 in IFN-non-responsive cancer cells show reduced anticancer activity by suppressing apoptosis via TRAIL and anti-apoptotic genes. Collectively, our study shows that IPS-1 induces anticancer activity through upregulation of pro-apoptotic gene TRAIL and downregulation of the anti-apoptotic genes BCL2, BIRC3 and PRKCE via IRF3 and IRF7 in type I IFN-dependent and -independent manners.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Interferon Tipo I/imunologia , Neoplasias/imunologia , Vírus da Doença de Newcastle/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Apoptose/imunologia , Proteína 3 com Repetições IAP de Baculovírus , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Regulação para Baixo , Células HEK293 , Humanos , Proteínas Inibidoras de Apoptose/biossíntese , Proteínas Inibidoras de Apoptose/genética , Fator Regulador 3 de Interferon/genética , Fator Regulador 7 de Interferon/genética , Invasividade Neoplásica/patologia , Poli I-C/imunologia , Proteína Quinase C-épsilon/biossíntese , Proteína Quinase C-épsilon/genética , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/genética , Regulação para Cima
8.
Anesteziol Reanimatol ; 60(6): 4-8, 2015.
Artigo em Russo | MEDLINE | ID: mdl-27025124

RESUMO

OBJECTIVE: To evaluate cardioprotective effects of remote ischemic preconditioning (RIPC) in cardiac surgery patients undergoing aortic valve replacement depending on the type of anesthesia and investigate the level of myocardial protein kinase C epsilon (PKC-ε) expression after RIPC. METHODS: In prospective randomized trial, forty eight patients aging from 50 to 75 years old (64 (56 ;69)) were included All patients were scheduled for aortic valve replacement using cardiopulmonary bypass (CPB). The patients were randomized into 4 groups: 1) RIPC applied during propofol anesthesia (RIPC prop, n = 12), 2) RIPC applied during sevoflurane anesthesia (RIPC sev, n = 12), 3) propofol anesthesia without RIPC (CONTROL prop, n = 12), 4) sevoflurane anesthesia without RIPC (CONTROL sev, n = 12). There was no difference found between the groups as to the baseline patient's data. RIPC protocol consisted of 3 simultaneous ischemic episodes of both lower limbs (5 minutes) with 5-min reperfusion intervals. PKC-ε expression in right atrial myocardium was assessed using Western blotting. Troponin I (cTnI) was estimated before anesthesia induction, after 30 min, 6, 12, 24, 48 hours after CPB completion. Also we calculated area under curve of cTnI (cTnI AUC). According to nonparametric distribution, data were assessed by the Mann-Whitney U-test and Newman-Keuls methodfor multigroup comparison. p < 0.05 was considered signifcant. The data are presented as median (25th; 75th percentile). RESULTS: Cardioprotective effects of RIPC were observed only after sevoflurane anesthesia: cTnI AUC was 134,8 (122,3; 232.4) ng/ml/48 h in CONTROL sev group and only 74.3 (64.7; 85.0) ng/ml/48 h in RIPC sev group (p < 0.05). RIPC applied during propofol anesthesia was not associated with cTnIAUC decrease: 93.8 (74.1; 246.8) ng/ml/48 h in CONTROL prop group and 122.5 (74.1; 185.0) ng/ml/48 h in RIPC prop group (p = 0.37). RIPC applied during sevoflurane anesthesia significantly increased PKC-ε expression: 1221 (921; 1438) U in CONTROL sev group vs 1882 (1564; 2131) U in RIPC sev group 6 (p < 0.05). RIPC implication during propofol anesthesia was not associated with any significant difference in PKC-ε expression in comparison with control group: 620 (436; 782) U in CONTROL prop group versus 788 (574;1063) U in RIPC prop group. In control groups, PKC-ε expression was significantly higher in sevoflurane anesthesia in comparison with propofol anesthesia. CONCLUSION: RIPC was only effective when it was applied during sevofiurane anesthesia. This was confirmed by PKC-ε expression increase and lower value of cTnI. There were no evidence of preconditioning and cardioprotection when MPG was initiated during propofol anesthesia.


Assuntos
Anestesia Geral/métodos , Valva Aórtica/cirurgia , Implante de Prótese de Valva Cardíaca/métodos , Precondicionamento Isquêmico/métodos , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Proteína Quinase C-épsilon/biossíntese , Idoso , Implante de Prótese de Valva Cardíaca/efeitos adversos , Humanos , Immunoblotting , Éteres Metílicos/administração & dosagem , Pessoa de Meia-Idade , Traumatismo por Reperfusão Miocárdica/etiologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/enzimologia , Propofol/administração & dosagem , Estudos Prospectivos , Proteína Quinase C-épsilon/metabolismo , Sevoflurano , Troponina I/sangue
9.
Exp Cell Res ; 330(2): 277-286, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25433270

RESUMO

RATIONALE: Vessel formation is a crucial event in tissue repair after injury. Thus, one assumption of innovative therapeutic approaches is the understanding of its molecular mechanisms. Notwithstanding our knowledge of the role of Protein Kinase C epsilon (PKCε) in cardio-protection and vascular restenosis, its role in vessel progenitor differentiation remains elusive. OBJECTIVE: Given the availability of PKCε pharmacological modulators already tested in clinical trials, the specific aim of this study is to unravel the role of PKCε in vessel progenitor differentiation, with implications in vascular pathology and vasculogenesis. METHODS AND RESULTS: Mouse Peri-Vascular Adipose Tissue (PVAT) was used as source of mesenchymal vessel progenitors. VEGF-induced differentiation of PVAT cells down-regulates both PKCε and p-PAK1 protein expression levels. PKCε overexpression and activation: i) reduced the expression levels of SMA and PECAM in endothelial differentiation of PVAT cells; ii) completely abrogated tubules formation in collagen gel assays; iii) increased the expression of p-PAK1. CONCLUSION: PKCε negatively interferes with vessel progenitor differentiation via interaction with PAK-1.


Assuntos
Tecido Adiposo/citologia , Células Endoteliais/citologia , Neovascularização Fisiológica/fisiologia , Proteína Quinase C-épsilon/metabolismo , Quinases Ativadas por p21/biossíntese , Actinas/biossíntese , Túnica Adventícia/citologia , Animais , Proteínas de Ligação ao Cálcio/biossíntese , Diferenciação Celular , Células Cultivadas , Reestenose Coronária/enzimologia , Regulação para Baixo , Ativação Enzimática , Camundongos , Proteínas dos Microfilamentos/biossíntese , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Proteína Quinase C-épsilon/biossíntese , Proteína Quinase C-épsilon/farmacologia , Proteínas Smad/biossíntese , Fator A de Crescimento do Endotélio Vascular/metabolismo , Calponinas
10.
Eur J Histochem ; 58(2): 2308, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24998921

RESUMO

PKCε, a DAG-dependent, Ca2+- independent kinase attenuates extent of fibrosis following tissue injury, suppresses apoptosis and promotes cell quiescence. In crescentic glomerulonephritis (CGN), glomerular epithelial cells (GEC) contribute to fibro-cellular crescent formation while they also transdifferentiate to a mesenchymal phenotype. The aim of this study was to assess PKCε expression in CGN. Using an antibody against PKC-ε phosphorylated at Ser729, we assessed its localization in rat model of immune-mediated rapidly progressive CGN. In glomeruli of control animals, pPKCε was undetectable. In animals with CGN, pPKCε was expressed exclusively in glomerular epithelial cells (GEC) and in GEC comprising fibrocellular crescents that had acquired a myofibroblast-type phenotype. In non-immune GEC injury induced by puromycin aminonucleoside and resulting in proteinuria of similar magnitude as in CGN, pPKCε expression was absent. There was constitutive pPKCε expression in distal convoluted tubules, collecting ducts and thick segments of Henley's loops in both control and experimental animals. We propose that pPKCε expression occurring in GEC and in fibrocellular crescentic lesions in CGN may facilitate PKCε dependent pathologic processes.


Assuntos
Regulação Enzimológica da Expressão Gênica , Glomerulonefrite/enzimologia , Glomérulos Renais/enzimologia , Proteína Quinase C-épsilon/biossíntese , Animais , Antimetabólitos Antineoplásicos/efeitos adversos , Antimetabólitos Antineoplásicos/farmacologia , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Glomerulonefrite/induzido quimicamente , Glomerulonefrite/patologia , Glomérulos Renais/patologia , Miofibroblastos/enzimologia , Miofibroblastos/patologia , Fosforilação/efeitos dos fármacos , Puromicina/efeitos adversos , Puromicina/farmacologia , Ratos , Ratos Sprague-Dawley , Serina/metabolismo
11.
J Biol Chem ; 289(28): 19823-38, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24825907

RESUMO

Overexpression of PKCϵ, a kinase associated with tumor aggressiveness and widely implicated in malignant transformation and metastasis, is a hallmark of multiple cancers, including mammary, prostate, and lung cancer. To characterize the mechanisms that control PKCϵ expression and its up-regulation in cancer, we cloned an ∼ 1.6-kb promoter segment of the human PKCϵ gene (PRKCE) that displays elevated transcriptional activity in cancer cells. A comprehensive deletional analysis established two regions rich in Sp1 and STAT1 sites located between -777 and -105 bp (region A) and -921 and -796 bp (region B), respectively, as responsible for the high transcriptional activity observed in cancer cells. A more detailed mutagenesis analysis followed by EMSA and ChIP identified Sp1 sites in positions -668/-659 and -269/-247 as well as STAT1 sites in positions -880/-869 and -793/-782 as the elements responsible for elevated promoter activity in breast cancer cells relative to normal mammary epithelial cells. RNAi silencing of Sp1 and STAT1 in breast cancer cells reduced PKCϵ mRNA and protein expression, as well as PRKCE promoter activity. Moreover, a strong correlation was found between PKCϵ and phospho-Ser-727 (active) STAT1 levels in breast cancer cells. Our results may have significant implications for the development of approaches to target PKCϵ and its effectors in cancer therapeutics.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Proteína Quinase C-épsilon/biossíntese , Elementos de Resposta , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição Sp1/metabolismo , Transcrição Gênica , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Proteínas de Neoplasias/genética , Proteína Quinase C-épsilon/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição Sp1/genética
12.
J Vis Exp ; (71)2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23328793

RESUMO

The protein kinase C (PKC) family of isozymes is involved in numerous physiological and pathological processes. Our recent data demonstrate that PKC regulates mitochondrial function and cellular energy status. Numerous reports demonstrated that the activation of PKC-a and PKC-ε improves mitochondrial function in the ischemic heart and mediates cardioprotection. In contrast, we have demonstrated that PKC-α and PKC-ε are involved in nephrotoxicant-induced mitochondrial dysfunction and cell death in kidney cells. Therefore, the goal of this study was to develop an in vitro model of renal cells maintaining active mitochondrial functions in which PKC isozymes could be selectively activated or inhibited to determine their role in regulation of oxidative phosphorylation and cell survival. Primary cultures of renal proximal tubular cells (RPTC) were cultured in improved conditions resulting in mitochondrial respiration and activity of mitochondrial enzymes similar to those in RPTC in vivo. Because traditional transfection techniques (Lipofectamine, electroporation) are inefficient in primary cultures and have adverse effects on mitochondrial function, PKC-ε mutant cDNAs were delivered to RPTC through adenoviral vectors. This approach results in transfection of over 90% cultured RPTC. Here, we present methods for assessing the role of PKC-ε in: 1. regulation of mitochondrial morphology and functions associated with ATP synthesis, and 2. survival of RPTC in primary culture. PKC-ε is activated by overexpressing the constitutively active PKC-ε mutant. PKC-ε is inhibited by overexpressing the inactive mutant of PKC-ε. Mitochondrial function is assessed by examining respiration, integrity of the respiratory chain, activities of respiratory complexes and F0F1-ATPase, ATP production rate, and ATP content. Respiration is assessed in digitonin-permeabilized RPTC as state 3 (maximum respiration in the presence of excess substrates and ADP) and uncoupled respirations. Integrity of the respiratory chain is assessed by measuring activities of all four complexes of the respiratory chain in isolated mitochondria. Capacity of oxidative phosphorylation is evaluated by measuring the mitochondrial membrane potential, ATP production rate, and activity of F0F1-ATPase. Energy status of RPTC is assessed by determining the intracellular ATP content. Mitochondrial morphology in live cells is visualized using MitoTracker Red 580, a fluorescent dye that specifically accumulates in mitochondria, and live monolayers are examined under a fluorescent microscope. RPTC viability is assessed using annexin V/propidium iodide staining followed by flow cytometry to determine apoptosis and oncosis. These methods allow for a selective activation/inhibition of individual PKC isozymes to assess their role in cellular functions in a variety of physiological and pathological conditions that can be reproduced in in vitro.


Assuntos
Rim/enzimologia , Rim/fisiologia , Mitocôndrias/fisiologia , Proteína Quinase C-alfa/biossíntese , Proteína Quinase C-épsilon/biossíntese , Trifosfato de Adenosina/biossíntese , Animais , Sobrevivência Celular/fisiologia , Células Cultivadas , Rim/citologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/enzimologia , Túbulos Renais Proximais/fisiologia , Mitocôndrias/enzimologia , ATPases Translocadoras de Prótons/metabolismo , Coelhos
13.
Am J Physiol Renal Physiol ; 301(1): F197-208, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21289057

RESUMO

PKC-ε activation mediates protection from ischemia-reperfusion injury in the myocardium. Mitochondria are a subcellular target of these protective mechanisms of PKC-ε. Previously, we have shown that PKC-ε activation is involved in mitochondrial dysfunction in oxidant-injured renal proximal tubular cells (RPTC; Nowak G, Bakajsova D, Clifton GL Am J Physiol Renal Physiol 286: F307-F316, 2004). The goal of this study was to examine the role of PKC-ε activation in mitochondrial dysfunction and to identify mitochondrial targets of PKC-ε in RPTC. The constitutively active and inactive mutants of PKC-ε were overexpressed in primary cultures of RPTC using the adenoviral technique. Increases in active PKC-ε levels were accompanied by PKC-ε translocation to mitochondria. Sustained PKC-ε activation resulted in decreases in state 3 respiration, electron transport rate, ATP production, ATP content, and activities of complexes I and IV and F(0)F(1)-ATPase. Furthermore, PKC-ε activation increased mitochondrial membrane potential and oxidant production and induced mitochondrial fragmentation and RPTC death. Accumulation of the dynamin-related protein in mitochondria preceded mitochondrial fragmentation. Antioxidants blocked PKC-ε-induced increases in the oxidant production but did not prevent mitochondrial fragmentation and cell death. The inactive PKC-ε mutant had no effect on mitochondrial functions, morphology, oxidant production, and RPTC viability. We conclude that active PKC-ε targets complexes I and IV and F(0)F(1)-ATPase in RPTC. PKC-ε activation mediates mitochondrial dysfunction, hyperpolarization, and fragmentation. It also induces oxidant generation and cell death, but oxidative stress is not the mechanism of RPTC death. These results show that in contrast to protective effects of PKC-ε activation in cardiomyocytes, sustained PKC-ε activation is detrimental to mitochondrial function and viability in RPTC.


Assuntos
Túbulos Renais Proximais/enzimologia , Túbulos Renais Proximais/fisiopatologia , Mitocôndrias/fisiologia , Proteína Quinase C-épsilon/metabolismo , Trifosfato de Adenosina/biossíntese , Adenoviridae/genética , Animais , Morte Celular/efeitos dos fármacos , Separação Celular , Sobrevivência Celular , Células Cultivadas , Ativação Enzimática/fisiologia , Feminino , Túbulos Renais Proximais/citologia , Cinética , L-Lactato Desidrogenase/metabolismo , Potenciais da Membrana/fisiologia , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/fisiologia , Consumo de Oxigênio/fisiologia , Proteína Quinase C-épsilon/biossíntese , Proteína Quinase C-épsilon/genética , ATPases Translocadoras de Prótons/metabolismo , Coelhos , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transfecção/métodos
14.
Int J Oncol ; 37(3): 719-29, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20664941

RESUMO

Tumor oxygen status is considered as a prognostic marker that impacts on malignant progression and outcome of tumor therapy. TNF-related apoptosis inducing ligand (TRAIL) plays a key role in cancer immunity, with potential applications in cancer therapy. Protein kinase C (PKC)epsilon, a transforming oncogene, has a role in the protection of cardiomyocytes and neurons from hypoxia-induced damage while, it can also modulate the susceptibility of tumor cells to TRAIL-induced cell death. Here we demonstrate that hypoxia induces a tumor cell phenotype highly sensitive to the cytotoxic effects of TRAIL. Based on the observation that: i) PKCepsilon expression levels are impaired during hypoxia, ii) the overexpression of PKCepsilon, but not of a kinase-inactive PKCepsilon mutant, is able to revert the hypoxia-induced sensitivity to TRAIL, iii) the down-modulation of PKCepsilon levels by RNA interference, on the contrary, induces the highly TRAIL-sensitive phenotype, iv) the inhibition of hypoxia-inducible transcription factor-1alpha (HIF-1alpha) by specific siRNA blocks both the hypoxia-induced down-modulation of PKCepsilon and the induction of the highly TRAIL-sensitive phenotype; we conclude that the HIF-1alpha upregulation during hypoxia is associated to PKCepsilon down-modulation that likely represents the key molecular event promoting the apoptogenic effects of TRAIL in hypoxic tumor cells.


Assuntos
Neoplasias/metabolismo , Proteína Quinase C-épsilon/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Regulação para Baixo , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células Jurkat , Células K562 , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Fenótipo , Prognóstico , Proteína Quinase C-épsilon/biossíntese , Proteína Quinase C-épsilon/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transfecção , Proteína bcl-X/biossíntese , Proteína bcl-X/genética
15.
Mol Cancer ; 9: 76, 2010 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-20398285

RESUMO

BACKGROUND: Protein kinase C (PKC) isoforms are potential targets for breast cancer therapy. This study was designed to evaluate which PKC isoforms might be optimal targets for different breast cancer subtypes. RESULTS: In two cohorts of primary breast cancers, PKCalpha levels correlated to estrogen and progesterone receptor negativity, tumor grade, and proliferative activity, whereas PKCdelta and PKCepsilon did not correlate to clinicopathological parameters. Patients with PKCalpha-positive tumors showed poorer survival than patients with PKCalpha-negative tumors independently of other factors. Cell line studies demonstrated that PKCalpha levels are high in MDA-MB-231 and absent in T47D cells which proliferated slower than other cell lines. Furthermore, PKCalpha silencing reduced proliferation of MDA-MB-231 cells. PKCalpha inhibition or downregulation also reduced cell migration in vitro. CONCLUSIONS: PKCalpha is a marker for poor prognosis of breast cancer and correlates to and is important for cell functions associated with breast cancer progression.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteína Quinase C-alfa/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Western Blotting , Neoplasias da Mama/metabolismo , Feminino , Imunofluorescência , Expressão Gênica , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Microscopia Confocal , Pessoa de Meia-Idade , Prognóstico , Proteína Quinase C-alfa/genética , Proteína Quinase C-delta/biossíntese , Proteína Quinase C-delta/genética , Proteína Quinase C-épsilon/biossíntese , Proteína Quinase C-épsilon/genética , RNA Interferente Pequeno , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/genética , Receptores de Progesterona/biossíntese , Receptores de Progesterona/genética , Transfecção
16.
Biochem Biophys Res Commun ; 390(4): 1377-81, 2009 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-19895783

RESUMO

Obestatin, the ghrelin-associated peptide, showed to activate MAPK signaling with no effect on Akt nor cell proliferating activity in rat tumor somatotroph cells (growth cells, GC). A sequential analysis of the obestatin transmembrane signaling pathway indicated a route involving the consecutive activation of G(i), PI3k, novel PKCepsilon, and Src for ERK1/2 activation. Furthermore, obestatin treatment triggers growth hormone (GH) release in the first 30min, being more acute at 15min. At 1h, obestatin treated cells showed the same levels in GH secretion than controls. Added to this functionality, obestatin was secreted by GC cells. Based on the capacity to stimulate GH release from somatotroph cells, obestatin may act directly in the pituitary through an autocrine/paracrine mechanism.


Assuntos
Grelina/farmacologia , Hormônio do Crescimento/metabolismo , Somatotrofos/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Ativação Enzimática , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/biossíntese , Fosfatidilinositol 3-Quinases/biossíntese , Proteína Quinase C-épsilon/biossíntese , Ratos , Somatotrofos/enzimologia , Somatotrofos/metabolismo , Quinases da Família src/biossíntese
17.
Cancer Res ; 69(6): 2287-95, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19244118

RESUMO

Limited information is available concerning the expression and role of microRNAs in prostate cancer. In this study, we investigated the involvement of miR-205 in prostate carcinogenesis. Significantly lower miR-205 expression levels were found in cancer than in normal prostate cell lines as well as in tumor compared with matched normal prostate tissues, with a particularly pronounced reduction in carcinomas from patients with local-regionally disseminated disease. Restoring the expression of miR-205 in prostate cancer cells resulted in cell rearrangements consistent with a mesenchymal-to-epithelial transition, such as up-regulation of E-cadherin and reduction of cell locomotion and invasion, and in the down-regulation of several oncogenes known to be involved in disease progression (i.e., interleukin 6, caveolin-1, EZH2). Our evidence suggests that these events are driven by the concurrent repression of specific predicted miR-205 targets, namely N-chimaerin, ErbB3, E2F1, E2F5, ZEB2, and protein kinase Cepsilon. Strikingly, the latter seemed to play a direct role in regulating epithelial-to-mesenchymal transition. In fact, its down-regulation led to a cell phenotype largely reminiscent of that of cells ectopically expressing miR-205. Overall, we showed for the first time that miR-205 exerts a tumor-suppressive effect in human prostate by counteracting epithelial-to-mesenchymal transition and reducing cell migration/invasion, at least in part through the down-regulation of protein kinase Cepsilon.


Assuntos
Genes Supressores de Tumor , MicroRNAs/genética , Neoplasias da Próstata/genética , Proteína Quinase C-épsilon/genética , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Regulação para Baixo , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Masculino , Mesoderma/patologia , MicroRNAs/biossíntese , Invasividade Neoplásica , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Proteína Quinase C-épsilon/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Homeobox 2 de Ligação a E-box com Dedos de Zinco
18.
Nat Cell Biol ; 10(8): 891-901, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18604201

RESUMO

The cell cycle is exquisitely controlled by multiple sequential regulatory inputs to ensure fidelity. Here we demonstrate that the final step in division, the physical separation of daughter cells, is controlled by a member of the PKC gene superfamily. Specifically, we have identified three phosphorylation sites within PKCepsilon that control its association with 14-3-3. These phosphorylations are executed by p38 MAP kinase (Ser 350), GSK3 (Ser 346) and PKC itself (Ser 368). Integration of these signals is essential during mitosis because mutations that prevent phosphorylation of PKCepsilon and/or PKCepsilon binding to 14-3-3 also cause defects in the completion of cytokinesis. Using chemical genetic and dominant-negative approaches it is shown that selective inhibition of PKCepsilon halts cells at the final stages of separation. This arrest is associated with persistent RhoA activation at the midbody and a delay in actomyosin ring dissociation. This study therefore identifies a new regulatory mechanism that controls exit from cytokinesis, which has implications for carcinogenesis.


Assuntos
Citocinese , Proteína Quinase C-épsilon/biossíntese , Proteínas 14-3-3/metabolismo , Animais , Linhagem Celular , Humanos , Camundongos , Fosforilação , Proteína Quinase C-épsilon/antagonistas & inibidores , Proteína Quinase C-épsilon/metabolismo , Transdução de Sinais , Transfecção , Proteína rhoA de Ligação ao GTP/metabolismo
19.
Cancer Res ; 67(18): 8828-38, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17875724

RESUMO

Prostate cancer is the most common type of cancer in men and ranks second only to lung cancer in cancer-related deaths. The management of locally advanced prostate cancer is difficult because the cancer often becomes hormone insensitive and unresponsive to current chemotherapeutic agents. Knowledge about the regulatory molecules involved in the transformation to androgen-independent prostate cancer is essential for the rational design of agents to prevent and treat prostate cancer. Protein kinase Cepsilon (PKCepsilon), a member of the novel PKC subfamily, is linked to the development of androgen-independent prostate cancer. PKCepsilon expression levels, as determined by immunohistochemistry of human prostate cancer tissue microarrays, correlated with the aggressiveness of prostate cancer. The mechanism by which PKCepsilon mediates progression to prostate cancer remains elusive. We present here for the first time that signal transducers and activators of transcription 3 (Stat3), which is constitutively activated in a wide variety of human cancers, including prostate cancer, interacts with PKCepsilon. The interaction of PKCepsilon with Stat3 was observed in human prostate cancer, human prostate cancer cell lines (LNCaP, DU145, PC3, and CW22rv1), and prostate cancer that developed in transgenic adenocarcinoma of mouse prostate mice. In reciprocal immunoprecipitation/blotting experiments, prostatic Stat3 coimmunoprecipitated with PKCepsilon. Localization of PKCepsilon with Stat3 was confirmed by double immunofluorescence staining. The interaction of PKCepsilon with Stat3 was PKCepsilon isoform specific. Inhibition of PKCepsilon protein expression in DU145 cells using specific PKCepsilon small interfering RNA (a) inhibited Stat3Ser727 phosphorylation, (b) decreased both Stat3 DNA-binding and transcriptional activity, and (c) decreased DU145 cell invasion. These results indicate that PKCepsilon activation is essential for constitutive activation of Stat3 and prostate cancer progression.


Assuntos
Neoplasias da Próstata/metabolismo , Proteína Quinase C-épsilon/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Linhagem Celular Tumoral , Citocinas/biossíntese , Citocinas/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica , Fosforilação , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteína Quinase C-épsilon/biossíntese , Proteína Quinase C-épsilon/genética , Fator de Transcrição STAT3/biossíntese , Fator de Transcrição STAT3/genética , Serina/metabolismo , Transdução de Sinais , Transcrição Gênica , Transfecção
20.
Cancer Res ; 67(13): 6053-63, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17616661

RESUMO

The protein kinase C (PKC) family of proteins plays important roles in growth regulation and is implicated in tumorigenesis. It has become clear that the role of PKC in tumorigenesis is cell context dependent and/or isoform specific. In this study, we showed for the first time by immunohistochemistry that overexpression of PKC epsilon was detected in the vast majority (>90%) of primary human non-small cell lung cancers (NSCLC) compared with normal lung epithelium. Inhibition of the PKC epsilon pathway using a kinase-inactive, dominant-negative PKC epsilon, PKC epsilon(KR), led to a significant inhibition of proliferation and anchorage-independent growth of human NSCLC cells in a p53-independent manner. This was accompanied by a specific induction of the cyclin-dependent kinase (cdk) inhibitor p21/Cip1 but not p27/Kip1. In response to serum stimulation, PKC epsilon(KR)-expressing cells showed a prolonged G(1)-S transition and delayed and reduced activation of cdk2 complexes, which was likely attributed to the increased binding of p21/Cip1 to cdk2. Furthermore, inhibition of PKC epsilon function either by expressing PKC epsilon(KR) or by small interfering RNA (siRNA)-mediated gene knockdown resulted in c-Myc down-regulation, which, in turn, regulated p21/Cip1 expression. Knockdown of PKC epsilon or c-Myc expression using siRNA led to induction of p21/Cip1 and attenuation of G(1)-S transition in NSCLC cells. Using p21(+/+) and p21(-/-) HCT116 isogenic cell lines, we further showed that growth inhibition by PKC epsilon(KR) required the function of p21/Cip1. Collectively, these results reveal an important role for PKC epsilon signaling in lung cancer and suggest that one potential mechanism by which PKC epsilon exerts its oncogenic activity is through deregulation of the cell cycle via a p21/Cip1-dependent mechanism.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Proteína Quinase C-épsilon/biossíntese , Idoso , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Proliferação de Células , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina , Feminino , Humanos , Neoplasias Pulmonares/genética , Masculino , Pessoa de Meia-Idade , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA