Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Exp Neurol ; 340: 113686, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33713658

RESUMEN

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.


Asunto(s)
Agonistas del Receptor de Adenosina A3/uso terapéutico , Adenosina/análogos & derivados , Aldehído Deshidrogenasa Mitocondrial/biosíntesis , Proteína Quinasa C-epsilon/biosíntesis , Receptor de Adenosina A3/biosíntesis , Hemorragia Subaracnoidea/tratamiento farmacológico , Tionucleósidos/uso terapéutico , Adenosina/farmacología , Adenosina/uso terapéutico , Agonistas del Receptor de Adenosina A3/farmacología , Animales , Relación Dosis-Respuesta a Droga , Inflamación/metabolismo , Inflamación/prevención & control , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Hemorragia Subaracnoidea/metabolismo , Tionucleósidos/farmacología
2.
Naunyn Schmiedebergs Arch Pharmacol ; 393(12): 2495-2506, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32157348

RESUMEN

Cardiomyocyte hypertrophy is a fatal factor in heart disease resulting in heart failure and even mortality. Although many studies have been focusing on the pathogenesis of cardiomyocyte hypertrophy, the exact molecular mechanisms are still unexclusive. In this study, we first found that the expression level of lncRNA Tincr was significantly decreased in the myocardial tissues of TAC mouse models of cardiomyocyte hypertrophy, and this result was further confirmed in H9C2 cells, a widely used rat myoblast cell lines. More intriguingly, we demonstrated that the aberration of Tincr is essential to the pathogenesis of cardiomyocyte hypertrophy, indicated by the re-induction of Tincr improving the heart functions of hypertrophic mice. In mechanism, we identified miR-31-5p as a direct target of Tincr using a widely used online bioinformatics tool StarBase, and this result was further experimentally validated using dual-luciferase reporter assay and real-time PCR. Also, we identified PRKCE as a direct target of miR-31-5p, and loss function of miR-31-5p significantly blocks the positive regulatory effect of Tincr on PRKCE expression in H9C2 cells. The knockdown of Tincr resulted in increased cardiomyocyte size, and, however, inhibition of miR-31-5p or overexpression of PRKCE significantly reversed the increased cardiomyocyte size. Taken together, our study showed that a novel Tincr-miR-31-5p axis targeting PRKCE was involved in cardiomyocyte hypertrophy, indicating that it may provide potential therapy in cardiomyocyte hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Aumento de la Célula , MicroARNs/biosíntesis , Miocitos Cardíacos/metabolismo , Proteína Quinasa C-epsilon/biosíntesis , ARN Largo no Codificante/biosíntesis , Animales , Secuencia de Bases , Cardiomegalia/genética , Cardiomegalia/patología , Línea Celular , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Miocitos Cardíacos/patología , Proteína Quinasa C-epsilon/genética , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , Ratas
3.
Basic Res Cardiol ; 112(6): 60, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28887629

RESUMEN

The cardioprotection of protein kinase Cepsilon (PKCε) against myocardial infarction (MI) mediated by its anti-apoptotic property and underlying mechanism of targeted regulation by microRNA (miRNA) are not established. MI-induced injury, PKCε expression, and targeted regulation of miRNA-143 (miR-143) to PKCε have been evaluated using animal MI and cellular hypoxic models conjugated with series of state-of-art molecular techniques. The results demonstrated that PKCε significantly downregulated along with increased infarcted area and apoptotic and necrotic damage in MI model, and the targeted relationship and potential binding profile were established between miR-143 and PKCε. Both in vivo and in vitro ischemic tests showed that miR-143 induced apoptosis and necrosis, which was reversed by antagomiR-143 or AMO-143. The upregulation of miR-143 by transfection of miR-143 in vitro also induced cell loss, and this effect of miR-143 was completely reversed by co-transfection of miR-143 with AMO-143. The identically deleterious action of miR-143 on mitochondrial membrane potential and ATP synthesis was also observed in both animal MI and cellular hypoxic models, as well as miR-143 overexpressed models and converted by either antagomiR or AMO. Importantly, overexpression of miR-143 downregulated PKCε in all tested models and this downregulation was reversed in the presence of antagomiR or AMO. The direct targeted regulation of miR-143 on PKCε was confirmed by luciferase reporter and miRNA-masking tests. In conclusion, MI-mediated upregulation of miR-143 inhibits PKCε expression and consequently interference with the cardioprotection of PKCε to mitochondrial, and leads to mitochondrial membrane potential dissipation and myocardial death eventually.


Asunto(s)
Regulación de la Expresión Génica/fisiología , MicroARNs/metabolismo , Isquemia Miocárdica/metabolismo , Proteína Quinasa C-epsilon/biosíntesis , Animales , Apoptosis/fisiología , Ratones , Mitocondrias/metabolismo , Isquemia Miocárdica/patología , Ratas , Ratas Sprague-Dawley
4.
Cell Death Dis ; 8(5): e2770, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28492560

RESUMEN

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.


Asunto(s)
Desoxicitidina/análogos & derivados , Neoplasias de la Vesícula Biliar , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , MicroARNs , Proteínas de Neoplasias , Proteína Quinasa C-epsilon , ARN Neoplásico , Regulación hacia Arriba/efectos de los fármacos , Subfamilia B de Transportador de Casetes de Unión a ATP/biosíntesis , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Desoxicitidina/farmacología , Femenino , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/metabolismo , Neoplasias de la Vesícula Biliar/patología , Humanos , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteína Quinasa C-epsilon/biosíntesis , Proteína Quinasa C-epsilon/genética , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Gemcitabina
5.
Cell Rep ; 19(2): 375-388, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28402859

RESUMEN

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.


Asunto(s)
Quimiocina CXCL13/genética , Fosfohidrolasa PTEN/biosíntesis , Neoplasias de la Próstata/genética , Proteína Quinasa C-epsilon/biosíntesis , Receptores CXCR5/genética , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Movimiento Celular , Quimiocina CXCL13/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , FN-kappa B , Metástasis de la Neoplasia , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/patología , Proteína Quinasa C-epsilon/genética , Receptores CXCR5/biosíntesis , Transducción de Señal
6.
PLoS One ; 11(2): e0150557, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26918336

RESUMEN

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.


Asunto(s)
Acetilcisteína/uso terapéutico , Antioxidantes/uso terapéutico , Isquemia Miocárdica/etiología , Nicotina/toxicidad , Efectos Tardíos de la Exposición Prenatal , Disfunción Ventricular Izquierda/etiología , Acetilcisteína/administración & dosificación , Animales , Antioxidantes/administración & dosificación , Circulación Coronaria/efectos de los fármacos , Susceptibilidad a Enfermedades , Evaluación Preclínica de Medicamentos , Femenino , Feto/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Bombas de Infusión Implantables , Masculino , Modelos Biológicos , Isquemia Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/prevención & control , Nicotina/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Fenotipo , Fosforilación/efectos de los fármacos , Embarazo , Proteína Quinasa C-epsilon/biosíntesis , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Recuperación de la Función , Disfunción Ventricular Izquierda/prevención & control
7.
Carcinogenesis ; 37(1): 72-80, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26586792

RESUMEN

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.


Asunto(s)
Carcinoma de Células Escamosas/prevención & control , Proteína Quinasa C-epsilon/genética , Neoplasias Cutáneas/prevención & control , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , 9,10-Dimetil-1,2-benzantraceno , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/genética , Carcinogénesis/efectos de la radiación , Carcinógenos , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa C-epsilon/biosíntesis , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Acetato de Tetradecanoilforbol , Rayos Ultravioleta
8.
Am J Physiol Heart Circ Physiol ; 310(4): H516-23, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26683901

RESUMEN

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.


Asunto(s)
Estimulantes del Sistema Nervioso Central/toxicidad , Metanfetamina/toxicidad , Isquemia Miocárdica/patología , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Peso al Nacer/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Femenino , Técnicas In Vitro , Masculino , Actividad Motora/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/patología , Isquemia Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/patología , Proteína Oncogénica v-akt/metabolismo , Fosforilación/efectos de los fármacos , Embarazo , Proteína Quinasa C-epsilon/biosíntesis , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales
9.
Cell Death Dis ; 6: e1758, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25950488

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Interferón Tipo I/inmunología , Neoplasias/inmunología , Virus de la Enfermedad de Newcastle/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Apoptosis/inmunología , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Regulación hacia Abajo , Células HEK293 , Humanos , Proteínas Inhibidoras de la Apoptosis/biosíntesis , Proteínas Inhibidoras de la Apoptosis/genética , Factor 3 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/genética , Invasividad Neoplásica/patología , Poli I-C/inmunología , Proteína Quinasa C-epsilon/biosíntesis , Proteína Quinasa C-epsilon/genética , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba
10.
Physiol Res ; 64(2): 191-201, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25317680

RESUMEN

Continuous normobaric hypoxia (CNH) renders the heart more tolerant to acute ischemia/reperfusion injury. Protein kinase C (PKC) is an important component of the protective signaling pathway, but the contribution of individual PKC isoforms under different hypoxic conditions is poorly understood. The aim of this study was to analyze the expression of PKCepsilon after the adaptation to CNH and to clarify its role in increased cardiac ischemic tolerance with the use of PKCepsilon inhibitory peptide KP-1633. Adult male Wistar rats were exposed to CNH (10 % O(2), 3 weeks) or kept under normoxic conditions. The protein level of PKCepsilon and its phosphorylated form was analyzed by Western blot in homogenate, cytosolic and particulate fractions; the expression of PKCepsilon mRNA was measured by RT-PCR. The effect of KP-1633 on cell viability and lactate dehydrogenase (LDH) release was analyzed after 25-min metabolic inhibition followed by 30-min re-energization in freshly isolated left ventricular myocytes. Adaptation to CNH increased myocardial PKCepsilon at protein and mRNA levels. The application of KP-1633 blunted the hypoxia-induced salutary effects on cell viability and LDH release, while control peptide KP-1723 had no effect. This study indicates that PKCepsilon is involved in the cardioprotective mechanism induced by CNH.


Asunto(s)
Adaptación Fisiológica/genética , Hipoxia/genética , Hipoxia/fisiopatología , Proteína Quinasa C-epsilon/genética , Animales , Supervivencia Celular/efectos de los fármacos , Hipoxia/enzimología , L-Lactato Deshidrogenasa/metabolismo , Masculino , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Proteína Quinasa C-epsilon/biosíntesis , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar
11.
Exp Cell Res ; 330(2): 277-286, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25433270

RESUMEN

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.


Asunto(s)
Tejido Adiposo/citología , Células Endoteliales/citología , Neovascularización Fisiológica/fisiología , Proteína Quinasa C-epsilon/metabolismo , Quinasas p21 Activadas/biosíntesis , Actinas/biosíntesis , Adventicia/citología , Animales , Proteínas de Unión al Calcio/biosíntesis , Diferenciación Celular , Células Cultivadas , Reestenosis Coronaria/enzimología , Regulación hacia Abajo , Activación Enzimática , Ratones , Proteínas de Microfilamentos/biosíntesis , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Proteína Quinasa C-epsilon/biosíntesis , Proteína Quinasa C-epsilon/farmacología , Proteínas Smad/biosíntesis , Factor A de Crecimiento Endotelial Vascular/metabolismo , Calponinas
12.
Anesteziol Reanimatol ; 60(6): 4-8, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-27025124

RESUMEN

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.


Asunto(s)
Anestesia General/métodos , Válvula Aórtica/cirugía , Implantación de Prótesis de Válvulas Cardíacas/métodos , Precondicionamiento Isquémico/métodos , Daño por Reperfusión Miocárdica/prevención & control , Proteína Quinasa C-epsilon/biosíntesis , Anciano , Implantación de Prótesis de Válvulas Cardíacas/efectos adversos , Humanos , Immunoblotting , Éteres Metílicos/administración & dosificación , Persona de Mediana Edad , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/enzimología , Propofol/administración & dosificación , Estudios Prospectivos , Proteína Quinasa C-epsilon/metabolismo , Sevoflurano , Troponina I/sangre
13.
Eur J Histochem ; 58(2): 2308, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24998921

RESUMEN

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.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Glomerulonefritis/enzimología , Glomérulos Renales/enzimología , Proteína Quinasa C-epsilon/biosíntesis , Animales , Antimetabolitos Antineoplásicos/efectos adversos , Antimetabolitos Antineoplásicos/farmacología , Células Epiteliales/enzimología , Células Epiteliales/patología , Glomerulonefritis/inducido químicamente , Glomerulonefritis/patología , Glomérulos Renales/patología , Miofibroblastos/enzimología , Miofibroblastos/patología , Fosforilación/efectos de los fármacos , Puromicina/efectos adversos , Puromicina/farmacología , Ratas , Ratas Sprague-Dawley , Serina/metabolismo
14.
J Biol Chem ; 289(28): 19823-38, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24825907

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Proteína Quinasa C-epsilon/biosíntesis , Elementos de Respuesta , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción Sp1/metabolismo , Transcripción Genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Proteínas de Neoplasias/genética , Proteína Quinasa C-epsilon/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción Sp1/genética
15.
J Vis Exp ; (71)2013 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-23328793

RESUMEN

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.


Asunto(s)
Riñón/enzimología , Riñón/fisiología , Mitocondrias/fisiología , Proteína Quinasa C-alfa/biosíntesis , Proteína Quinasa C-epsilon/biosíntesis , Adenosina Trifosfato/biosíntesis , Animales , Supervivencia Celular/fisiología , Células Cultivadas , Riñón/citología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/fisiología , Mitocondrias/enzimología , ATPasas de Translocación de Protón/metabolismo , Conejos
16.
Biochem Biophys Res Commun ; 426(2): 237-41, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22935420

RESUMEN

UNLABELLED: Pervious biochemical and hemodynamic studies have highlighted the important role of εPKC in cardioprotection during ischemic preconditioning. However, little is known about the electrophysiological consequences of εPKC modulation in ischemic hearts. Membrane permeable peptide εPKC selective activator and inhibitor were used to investigate the role of εPKC modulation in reperfusion arrhythmias. METHODS: Protein transduction domain from HIV-TAT was used as a carrier for peptide delivery into intact Langendorff perfused guinea pig hearts. Action potentials were imaged and mapped (124 sites) using optical techniques and surface ECG was continuously recorded. Hearts were exposed to 30 min stabilization period, 15 min of no-flow ischemia, followed by 20 min reperfusion. Peptides (0.5 µM) were infused as follows: (a) control (vehicle-TAT peptide; TAT-scrambled ψεRACK peptide); (b) εPKC agonist (TAT-ψεRACK); (c) εPKC antagonist (TAT-εV1). RESULTS: Hearts treated with εPKC agonist ψεRACK had reduced incidence of ventricular tachycardia (VT, 64%) and fibrillation (VF, 50%) compared to control (VT, 80%, P<0.05) and (VF, 70%, P < 0.05). However, the highest incidence of VT (100%, P < 0.05) and VF (80%) occurred in hearts treated with εPKC antagonist peptide εV1 compared to control and to εPKC agonist ψεRACK. Interestingly, at 20 min reperfusion, 100% of hearts treated with εPKC agonist ψεRACK exhibited complete recovery of action potentials compared to 40% (P < 0.05) of hearts treated with εPKC antagonist peptide, εV1 and 65% (P < 0.5) of hearts in control. At 20 min reperfusion, maps of action potential duration from εPKC agonist ψεRACK showed minimal dispersion (48.2 ± 9 ms) compared to exacerbated dispersion (115.4 ± 42 ms, P < 0.05) in εPKC antagonist and control (67 ± 20 ms, P<0.05). VT/VF and dispersion from hearts treated with scrambled agonist or antagonist peptides were similar to control. CONCLUSION: The results demonstrate that εPKC activation by ψεRACK peptide protects intact hearts from reperfusion arrhythmias and affords better recovery. On the other hand, inhibition of εPKC increased the incidence of arrhythmias and worsened recovery compared to controls. The results carry significant therapeutic implications for the treatment of acute ischemic heart disease by preconditioning-mimicking agents.


Asunto(s)
Arritmias Cardíacas/enzimología , Daño por Reperfusión Miocárdica/complicaciones , Proteína Quinasa C-epsilon/biosíntesis , Animales , Arritmias Cardíacas/etiología , Activación Enzimática , Cobayas , Técnicas In Vitro
17.
Exp Lung Res ; 38(8): 383-95, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22897707

RESUMEN

Organic dust samples from swine confinement facilities elicit pro-inflammatory cytokine/chemokine release from bronchial epithelial cells and monocytes, dependent, in part, upon dust-induced activation of the protein kinase C (PKC) isoform, PKCε. PKCε is also rapidly activated in murine tracheal epithelial cells following in vivo organic dust challenges, yet the functional role of PKCε in modulating dust-induced airway inflammatory outcomes is not defined. Utilizing an established intranasal inhalation animal model, experiments investigated the biologic and physiologic responses following organic dust extract (ODE) treatments in wild-type (WT) and PKCε knock-out (KO) mice. We found that neutrophil influx increased more than twofold in PKCε KO mice following both a one-time challenge and 3 weeks of daily challenges with ODE as compared with WT mice. Lung pathology revealed increased bronchiolar and alveolar inflammation, lymphoid aggregates, and T cell influx in ODE-treated PKCε KO mice. Airway hyperresponsiveness to methacholine increased in PKCε KO + ODE to a greater magnitude than WT + ODE animals. There were no significant differences in cytokine/chemokine release elicited by ODE treatment between groups. However, ODE-induced nitric oxide (NO) production differed in that ODE exposure increased nitrate levels in WT mice but not in PKCε KO mice. Moreover, ODE failed to upregulate NO from ex vivo stimulated PKCε KO lung macrophages. Collectively, these studies demonstrate that PKCε-deficient mice were hypersensitive to organic dust exposure and suggest that PKCε is important in the normative lung inflammatory response to ODE. Dampening of ODE-induced NO may contribute to these enhanced inflammatory findings.


Asunto(s)
Hiperreactividad Bronquial/enzimología , Polvo/inmunología , Mediadores de Inflamación/metabolismo , Pulmón/efectos de los fármacos , Proteína Quinasa C-epsilon/biosíntesis , Animales , Hiperreactividad Bronquial/genética , Hiperreactividad Bronquial/inmunología , Pruebas de Provocación Bronquial , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Exposición a Riesgos Ambientales/efectos adversos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Femenino , Pulmón/metabolismo , Pulmón/patología , Masculino , Cloruro de Metacolina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Proteína Quinasa C-epsilon/deficiencia , Proteína Quinasa C-epsilon/genética , Linfocitos T/patología
18.
Brain Res ; 1461: 51-63, 2012 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-22578356

RESUMEN

The cervical facet joint and its capsule are a common source of neck pain from whiplash. Mechanical hyperalgesia elicited by painful facet joint distraction is associated with spinal neuronal hyperexcitability that can be induced by transmitter/receptor systems that potentiate the synaptic activation of neurons. This study investigated the temporal response of a glutamate receptor and transporters in the dorsal root ganglia (DRG) and spinal cord. Bilateral C6/C7 facet joint distractions were imposed in the rat either to produce behavioral sensitivity or without inducing any sensitivity. Neuronal metabotropic glutamate receptor-5 (mGluR5) and protein kinase C-epsilon (PKCε) expression in the DRG and spinal cord were evaluated on days 1 and 7. Spinal expression of a glutamate transporter, excitatory amino acid carrier 1 (EAAC1), was also quantified at both time points. Painful distraction produced immediate behavioral hypersensitivity that was sustained for 7 days. Increased expression of mGluR5 and PKCε in the DRG was not evident until day 7 and only following painful distraction; this increase was observed in small-diameter neurons. Only painful facet joint distraction produced a significant increase (p<0.001) in neuronal mGluR5 over time, and this increase also was significantly elevated (p≤0.05) over responses in the other groups at day 7. However, there were no differences in spinal PKCε expression on either day or between groups. Spinal EAAC1 expression was significantly increased (p<0.03) only in the nonpainful groups on day 7. Results from this study suggest that spinal glutamatergic plasticity is selectively modulated in association with facet-mediated pain.


Asunto(s)
Transportador 3 de Aminoácidos Excitadores/biosíntesis , Ganglios Espinales/metabolismo , Receptores de Glutamato Metabotrópico/biosíntesis , Médula Espinal/metabolismo , Lesiones por Latigazo Cervical/metabolismo , Articulación Cigapofisaria/metabolismo , Animales , Transportador 3 de Aminoácidos Excitadores/fisiología , Ganglios Espinales/patología , Masculino , Dolor/metabolismo , Dolor/patología , Dimensión del Dolor/métodos , Proteína Quinasa C-epsilon/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/fisiología , Médula Espinal/patología , Lesiones por Latigazo Cervical/patología , Articulación Cigapofisaria/patología
19.
J Invest Dermatol ; 132(8): 1988-97, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22475757

RESUMEN

Protein kinase C (PKC) isoforms have crucial roles in cutaneous signaling. Interestingly, we lack information about their involvement in human sebaceous gland biology. Therefore, in this current study, we investigated the functions of the PKC system in human immortalized SZ95 sebocytes. Using molecular biological approaches, imaging, and functional assays, we report that SZ95 sebocytes express the conventional cPKCα; the novel nPKCδ, ɛ, and η; and the atypical aPKCζ. Activation of the PKC system by phorbol 12-myristate 13-acetate (PMA) stimulated lipid synthesis (a hallmark of differentiation) and resulted in translocation and then downregulation of cPKCα and nPKCδ. In good accord with these findings, the effect of PMA was effectively abrogated by inhibitors and short interfering RNA-mediated "silencing" of cPKCα and nPKCδ. Of further importance, molecular or pharmacological inhibition of nPKCδ also prevented the lipogenic and apoptosis-promoting action of arachidonic acid. Finally, we also found that "knockdown" of the endogenous aPKCζ activity markedly increased basal lipid synthesis and apoptosis, suggesting its constitutive activity in suppressing these processes. Collectively, our findings strongly argue for the fact that certain PKCs have pivotal, isoform-specific, differential, and antagonistic roles in the regulation of human sebaceous gland-derived sebocyte biology.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Proteína Quinasa C-alfa/biosíntesis , Proteína Quinasa C-delta/biosíntesis , Proteína Quinasa C-epsilon/biosíntesis , Proteína Quinasa C/biosíntesis , Glándulas Sebáceas/citología , Apoptosis , Diferenciación Celular , Regulación hacia Abajo , Silenciador del Gen , Humanos , Lípidos/química , Necrosis , Isoformas de Proteínas , ARN Interferente Pequeño/metabolismo , Acetato de Tetradecanoilforbol/farmacología
20.
J Immunol ; 187(9): 4721-32, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21964026

RESUMEN

We have studied the functional role of protein kinase Cε (PKCε) in the control of human CD4(+) T cell proliferation and in their response to TGF-1ß. We demonstrate that PKCε sustains CD4(+) T cell proliferation triggered in vitro by CD3 stimulation. Transient knockdown of PKCε expression decreases IL-2R chain transcription, and consequently cell surface expression levels of CD25. PKCε silencing in CD4 T cells potentiates the inhibitory effects of TGF-1ß, whereas in contrast, the forced expression of PKCε virtually abrogates the inhibitory effects of TGF-1ß. Being that PKCε is therefore implicated in the response of CD4 T cells to both CD3-mediated proliferative stimuli and TGF-1ß antiproliferative signals, we studied it in Hashimoto thyroiditis (HT), a pathology characterized by abnormal lymphocyte proliferation and activation. When we analyzed CD4 T cells from HT patients, we found a significant increase of PKCε expression, accounting for their enhanced survival, proliferation, and decreased sensitivity to TGF-1ß. The increased expression of PKCε in CD4(+) T cells of HT patients, which is described for the first time, to our knowledge, in this article, viewed in the perspective of the physiological role of PKCε in normal Th lymphocytes, adds knowledge to the molecular pathophysiology of HT and creates potentially new pharmacological targets for the therapy of this disease.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Enfermedad de Hashimoto/enzimología , Enfermedad de Hashimoto/inmunología , Proteína Quinasa C-epsilon/fisiología , Factor de Crecimiento Transformador beta1/farmacología , Adulto , Animales , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Femenino , Enfermedad de Hashimoto/metabolismo , Humanos , Células Jurkat , Activación de Linfocitos/inmunología , Masculino , Ratones , Persona de Mediana Edad , Proteína Quinasa C-epsilon/biosíntesis , Proteína Quinasa C-epsilon/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA