Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Respir Cell Mol Biol ; 62(6): 732-746, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32048876

RESUMO

Pulmonary vasoconstriction resulting from intermittent hypoxia (IH) contributes to pulmonary hypertension (pHTN) in patients with sleep apnea (SA), although the mechanisms involved remain poorly understood. Based on prior studies in patients with SA and animal models of SA, the objective of this study was to evaluate the role of PKCß and mitochondrial reactive oxygen species (mitoROS) in mediating enhanced pulmonary vasoconstrictor reactivity after IH. We hypothesized that PKCß mediates vasoconstriction through interaction with the scaffolding protein PICK1 (protein interacting with C kinase 1), activation of mitochondrial ATP-sensitive potassium channels (mitoKATP), and stimulated production of mitoROS. We further hypothesized that this signaling axis mediates enhanced vasoconstriction and pHTN after IH. Rats were exposed to IH or sham conditions (7 h/d, 4 wk). Chronic oral administration of the antioxidant Tempol or the PKCß inhibitor LY-333531 abolished IH-induced increases in right ventricular systolic pressure and right ventricular hypertrophy. Furthermore, scavengers of O2- or mitoROS prevented enhanced PKCß-dependent vasoconstrictor reactivity to endothelin-1 in pulmonary arteries from IH rats. In addition, this PKCß/mitoROS signaling pathway could be stimulated by the PKC activator PMA in pulmonary arteries from control rats, and in both rat and human pulmonary arterial smooth muscle cells. These responses to PMA were attenuated by inhibition of mitoKATP or PICK1. Subcellular fractionation and proximity ligation assays further demonstrated that PKCß acutely translocates to mitochondria upon stimulation and associates with PICK1. We conclude that a PKCß/mitoROS signaling axis contributes to enhanced vasoconstriction and pHTN after IH. Furthermore, PKCß mediates pulmonary vasoconstriction through interaction with PICK1, activation of mitoKATP, and subsequent mitoROS generation.


Assuntos
Hipertensão Pulmonar/fisiopatologia , Hipóxia/fisiopatologia , Mitocôndrias/fisiologia , Proteína Quinase C beta/fisiologia , Artéria Pulmonar/fisiopatologia , Vasoconstrição/fisiologia , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Células Cultivadas , Óxidos N-Cíclicos/farmacologia , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/metabolismo , Sequestradores de Radicais Livres/farmacologia , Humanos , Hipertensão Pulmonar/etiologia , Hipóxia/complicações , Hipóxia/enzimologia , Indóis/farmacologia , Masculino , Maleimidas/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/fisiopatologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Canais de Potássio/metabolismo , Mapeamento de Interação de Proteínas , Artéria Pulmonar/enzimologia , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Síndromes da Apneia do Sono/fisiopatologia , Marcadores de Spin , Acetato de Tetradecanoilforbol/farmacologia
2.
Pharmacol Res ; 152: 104618, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31891789

RESUMO

Several decades have passed since resveratrol (RSV) was first identified in red wine. Researchers have reported the pleiotropic anti-oxidant, anti-inflammatory, anti-cancer, anti-aging, and neuronal protective effects of resveratrol and its glycosylated derivative. However, few studies have distinguished the minute differences in the properties between resveratrol and its glycosylated derivative in terms of synaptic plasticity. As an abundant natural product of glycosylated resveratrol, the derivative 2,3,4',5-tetrahydroxystilbene-2-O-ß-d-glucoside (TSG) has been determined to be a better option for long-term potentiation (LTP) in the hippocampus under physiological and pathological conditions than resveratrol. TSG, as well as its parent molecule RSV, could elicit early-LTP and recover fast excitatory postsynaptic potentials (EPSPs) in the hippocampus. Using various modalities, including pre- and post-whole-cell patch clamping techniques in the calyx of Held, pharmacological inhibition of the N-methyl-d-aspartic acid receptor (NMDAr) and the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAr) as well as protein kinase C (PKC) activation, we demonstrated that TSG, unlike RSV, could merely promote NMDA-mediated EPSC via PKCß cascade. Our results provide new knowledge that glycosylation of resveratrol could significantly improve its specificity in promoting sole NMDAr mediation of EPSPs, in addition to improving solubility and resistance against oxidation in vivo. These observations could contribute to further exploration of pharmaceutical evaluation of glycosylated stilbene in the future.


Assuntos
Glucosídeos/farmacologia , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Estilbenos/farmacologia , Animais , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Hipocampo/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Quinase C beta/fisiologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/fisiologia
3.
Cell Signal ; 64: 109418, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31525436

RESUMO

G Protein-activated K+ channels (GIRK) channels are inhibited by depletion of PtdIns(4,5)P2(PIP2), and/or channel phosphorylation by proteinkinase C (PKC). By using FRET-based biosensors, expressed in HEK293 cells or in atrial myocytes, we quantified receptor-specific Gq-coupled receptor (GqPCR) signalling on the level of phospholipase C (PLC) activation by monitoring PIP2-depletion and diacylglycerol (DAG) formation. Simultaneous voltage-clamp experiments on GIRK channel activity were performed as a functional readout for Gq-coupled α1B- and ET-receptor-induced signalling. GqPCR-induced fast inhibition of GIRK channel activity is mediated by depletion of PIP2, whereas phosphorylation of GIRK channels results in delayed, but effective GIRK current inhibition. We demonstrate a receptor-induced inhibitory component on GIRK activity that is independent of PIP2-depletion, but attributed to the activation of Ca2+-dependent PKC isoforms. As a novel finding, we demonstrate receptor-dependent differences in GIRK inhibition according to receptor-specific activation of the Ca2+-dependent PKC isoforms PKCα and PKCß. Pharmacological inhibition of PKCα, but not of PKCß, abolishes GIRK inhibition induced by stimulation of α1B-receptors. In contrast, ET-R-induced reduction of GIRK activity is sensitive to pharmacological block of PKCß, but not of PKCα. Coexpression of α1B-receptors (or ETB-R) and PKCα (or PKCß) in HEK 293 cells increased homologous receptor desensitization as indicated by a rapid decline of the CKAR FRET signal monitoring receptor activity. These data suggest that receptor-species dependent differences in PKC isoform activation regulate both GIRK channel activity and the strength of the receptor signal via a negative feedback mechanism.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Miócitos Cardíacos/metabolismo , Proteína Quinase C beta/fisiologia , Proteína Quinase C-alfa/fisiologia , Animais , Transferência Ressonante de Energia de Fluorescência/métodos , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Átrios do Coração , Humanos , Ratos , Receptores Adrenérgicos alfa 1/metabolismo
4.
Oncogene ; 38(1): 120-139, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30082911

RESUMO

Glioblastomas (GBM) are the most aggressive brain cancers without effective therapeutics. The Hippo pathway transcriptional coactivators YAP/TAZ were implicated as drivers in GBM progression and could be therapeutic targets. Here we found in an unbiased screen of 1650 compounds that amlodipine is able to inhibit survival of GBM cells by suppressing YAP/TAZ activities. Instead of its known function as an L-type calcium channel blocker, we found that amlodipine is able to activate Ca2+ entry by enhancing store-operated Ca2+ entry (SOCE). Amlodipine as well as approaches that cause store depletion and activate SOCE trigger phosphorylation and activation of Lats1/2, which in turn phosphorylate YAP/TAZ and prevent their accumulation in the cell nucleus. Furthermore, we identified that protein kinase C (PKC) beta II is a major mediator of Ca2+-induced Lats1/2 activation. Ca2+ induces accumulation of PKC beta II in an actin cytoskeletal compartment. Such translocation depends on inverted formin-2 (INF2). Depletion of INF2 disrupts both PKC beta II translocation and Lats1/2 activation. Functionally, we found that elevation of cytosolic Ca2+ or PKC beta II expression inhibits YAP/TAZ-mediated gene transcription. In vivo PKC beta II expression inhibits GBM tumor growth and prolongs mouse survival through inhibition of YAP/TAZ in an orthotopic mouse xenograft model. Our studies indicate that Ca2+ is a crucial intracellular cue that regulates the Hippo pathway and that triggering SOCE could be a strategy to target YAP/TAZ in GBM.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Anlodipino/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Glioblastoma/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Fosfoproteínas/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/antagonistas & inibidores , Aciltransferases , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Linhagem Celular Tumoral , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Técnicas de Silenciamento de Genes , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Via de Sinalização Hippo , Humanos , Ionomicina/farmacologia , Camundongos , Camundongos Nus , Proteínas de Neoplasias/fisiologia , Proteína ORAI1/antagonistas & inibidores , Proteína ORAI1/genética , Proteína ORAI1/fisiologia , Fosfoproteínas/genética , Fosforilação/efeitos dos fármacos , Proteína Quinase C beta/fisiologia , Proteínas Quinases/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , RNA Interferente Pequeno/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais/fisiologia , Tapsigargina/farmacologia , Fatores de Transcrição/genética , Proteínas de Sinalização YAP
5.
Acta Med Okayama ; 72(4): 359-367, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30140083

RESUMO

Abnormal glucose metabolism during pregnancy is an established risk factor for preeclampsia (PE). Disruption of the balance between placental angiogenic factors is linked to PE pathophysiology. We examined whether hypoxia-induced factor-1α (HIF-1α) and protein kinase Cß (PKCß) are involved in the regulation of placental angiogenic factors under high-glucose conditions in vitro. The human choriocarcinoma cell lines BeWo and JEG-3, and the human trophoblast cell line HTR-8/SVneo were cultured with 10 and 25 mmol/L glucose [control glucose group (CG) and high-glucose group (HG), respectively]. We examined the changes in HIF-1α, soluble fms-like tyrosine kinase-1 (sFlt-1), placental growth factor (PlGF), and vascular endothelial growth factor (VEGF) expression in the CG and HG by real-time PCR and ELISA. PKC activation was also measured by ELISA. The expressions of HIF-1α, sFlt-1, PlGF, and VEGF were significantly higher in the HG than in the CG. PKC activity was significantly increased in the HG. High glucose affected the expression of angiogenic factors in choriocarcinoma cells via the PKCß and HIF-1α pathways, suggesting their involvement in PE pathogenesis.


Assuntos
Glucose/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Placenta/metabolismo , Proteína Quinase C beta/fisiologia , Linhagem Celular Tumoral , Coriocarcinoma/patologia , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteínas de Membrana/genética , Gravidez , RNA Mensageiro/análise , Transdução de Sinais , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
6.
Adv Clin Exp Med ; 26(9): 1335-1342, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29442453

RESUMO

BACKGROUND: Arsenic trioxide (ATO) is a well-recognized antileukemic drug used for the treatment of newly diagnosed and relapsed acute promyelocytic leukemia (APL). A major drawback of therapy with ATO is the development of APL cell resistance, the mechanisms of which are still not clear. OBJECTIVES: The aim of this study was to investigate the role of the PI3K/Akt signaling pathway in ATOtreated human acute myeloid leukemia (HL-60) cells and in ATO-resistant clones. MATERIAL AND METHODS: The cytotoxicity of ATO was assessed using Trypan blue staining or a WST-1 reduction assay. The Akt phosphorylation level was measured by immunofluorescent staining and flow cytometry. Gene expression analysis was performed using real-time polymerase chain reaction (PCR). RESULTS: The clones derived by culturing for 8-12 weeks in the presence of 1.75, 2.5, and 5 µM ATO were characterized by high viability but a slower growth rate compared to the parental HL-60 cells. The flow cytometry analysis showed that in the parental cells the levels of p-Akt were undetectable or very low, and that ATO had no effect on the level of p-Akt in either the ATO-treated parental cells or the clones. The gene expression analysis revealed that some of the genes involved in the Akt pathway may play a key role in the induction of resistance to ATO, e.g., genes encoding cyclin D1 (CCND1), fork head box O1 (FOXO1), Jun oncogene (JUN), protein kinase C isoform B1 (PRKCB1), because their expression profiles were predominantly changed in the clones and/or the ATO-treated parental HL-60 cells. CONCLUSIONS: The overall results indicate that CCND1, FOXO1, and JUN may contribute to the induction of resistance to ATO, and that the C-Jun N-terminal kinase (JNK) signaling pathway may have greater significance than the phosphoinositide 3-kinase (PI3K)/Akt pathway in mediating the cytotoxic effects of ATO and the development of resistance to ATO in the HL-60 cell line.


Assuntos
Antineoplásicos/farmacologia , Perfilação da Expressão Gênica , Genes jun/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Trióxido de Arsênio , Arsenicais , Ciclina D1/fisiologia , Resistencia a Medicamentos Antineoplásicos , Proteína Forkhead Box O1/fisiologia , Células HL-60 , Humanos , Óxidos , Proteína Quinase C beta/fisiologia
7.
Br J Pharmacol ; 173(5): 870-87, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26660275

RESUMO

BACKGROUND AND PURPOSE: We investigated the hypothesis that elevated glucose increases contractile responses in vascular smooth muscle and that this enhanced constriction occurs due to the glucose-induced PKC-dependent inhibition of voltage-gated potassium channels. EXPERIMENTAL APPROACH: Patch-clamp electrophysiology in rat isolated mesenteric arterial myocytes was performed to investigate the glucose-induced inhibition of voltage-gated potassium (Kv ) current. To determine the effects of glucose in whole vessel, wire myography was performed in rat mesenteric, porcine coronary and human internal mammary arteries. KEY RESULTS: Glucose-induced inhibition of Kv was PKC-dependent and could be pharmacologically dissected using PKC isoenzyme-specific inhibitors to reveal a PKCß-dependent component of Kv inhibition dominating between 0 and 10 mM glucose with an additional PKCα-dependent component becoming evident at concentrations greater than 10 mM. These findings were supported using wire myography in all artery types used, where contractile responses to vessel depolarization and vasoconstrictors were enhanced by increasing bathing glucose concentration, again with evidence for distinct and complementary PKCα/PKCß-mediated components. CONCLUSIONS AND IMPLICATIONS: Our results provide compelling evidence that glucose-induced PKCα/PKCß-mediated inhibition of Kv current in vascular smooth muscle causes an enhanced constrictor response. Inhibition of Kv current causes a significant depolarization of vascular myocytes leading to marked vasoconstriction. The PKC dependence of this enhanced constrictor response may present a potential therapeutic target for improving microvascular perfusion following percutaneous coronary intervention after myocardial infarction in hyperglycaemic patients.


Assuntos
Vasos Coronários/efeitos dos fármacos , Glucose/farmacologia , Artéria Torácica Interna/efeitos dos fármacos , Artérias Mesentéricas/efeitos dos fármacos , Proteína Quinase C beta/fisiologia , Proteína Quinase C-alfa/fisiologia , Animais , Vasos Coronários/fisiologia , Humanos , Técnicas In Vitro , Isoenzimas/antagonistas & inibidores , Isoenzimas/fisiologia , Masculino , Artéria Torácica Interna/fisiologia , Artérias Mesentéricas/fisiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Proteína Quinase C beta/antagonistas & inibidores , Proteína Quinase C-alfa/antagonistas & inibidores , Ratos Wistar , Suínos , Vasoconstrição/efeitos dos fármacos
9.
Zhongguo Dang Dai Er Ke Za Zhi ; 17(3): 275-80, 2015 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-25815500

RESUMO

OBJECTIVE: To explore the roles of PKCß/P66Shc oxidative stress signal pathway in mediating hyperoxia-induced reactive oxgen species (ROS) production in alveolar epithelial cells (A549) and the protective effects of PKCß inhibitor on hyperoxia-induced injuries of alveolar epithelial cells. METHODS: A549 cells were cultured in vitro and randomly divided into three groups: control, hyperoxia and PKCß inhibitor LY333531 treatment. The hyperoxia group was exposed to a mixture of O2 (950 mL/L) and CO2 (50 mL/L) for 10 minutes and then cultured in a closed environment. The LY333531 group was treated with PKCß inhibitor LY333531 of 10 µmol/L for 24 hours before hyperoxia induction. Cells were collected 24 hours after culture and the levels of PKCß, Pin1, P66Shc and P66Shc-Ser36 were detected by Western blot. The intracellular translocation of P66Shc, the production of ROS and cellular mitochondria membrane potential were measured using the confocal microscopy. RESULTS: Compared with the control group, the levels of PKCß, Pin1, P66Shc and P-P66Shc-Ser36 in A549 cells 24 hours after culture increased significantly in the hyperoxia group. These changes in the hyperoxia group were accompanied with an increased translocation rate of P66Shc from cytoplasm into mitochondria, an increased production of mitochondrial ROS, and a reduced mitochondrial membrane potential. Compared with the hyperoxia group, the levels of Pin1, P66Shc and P66Shc-Ser36 in A549 cells, the translocation rate of P66Shc from cytoplasm into mitochondria and the production of mitochondrial ROS decreased significantly, while the mitochondrial membrane potential increased significantly in the LY333531 treatment group. However, there were significant differences in the above mentioned measurements between the LY333531 treatment and control groups. CONCLUSIONS: Hyperoxia can increase the expression of PKCß in alveolar epithelial cells and production of mitochondrial ROS and decrease mitochondrial membrane potential. PKCß inhibitor LY333531 can partially disrupt these changes and thus alleviate the hyperoxia-induced alveolar epithelial cell injury.


Assuntos
Hipóxia Celular , Estresse Oxidativo , Proteína Quinase C beta/fisiologia , Alvéolos Pulmonares/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Transdução de Sinais/fisiologia , Células Cultivadas , Células Epiteliais/metabolismo , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Alvéolos Pulmonares/citologia , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
10.
Anticancer Res ; 35(3): 1291-6, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25750277

RESUMO

BACKGROUND: The objective of this study was to examine the effect of specific Protein kinase C (PKC) isoform re-expression in solid malignancies, particularly head and neck squamous cell carcinoma cell lines, and the impact this may have on treatment with known activators of PKC. MATERIALS AND METHODS: The constitutive expression of PKC isoforms were determined in six head and neck squamous cell carcinoma (SCC) cell lines. Cytotoxicity of the prototypic phorbol ester, 12-O-tetradecanoylphorbol-13-acetate (TPA) and the novel diterpene ester PEP005 was established. Viral transduction to re-express PKCß isoforms in two of these cell lines was performed, and its effect on the sensitivity to the compounds was quantified. RESULTS: Tongue and hypopharyngeal SCC cell lines were resistant to both TPA and PEP005, with the concentration required to inhibit growth by 50% (IC50) being >1,000 ng/ml. CAL-27 (tongue SCC) and FaDu (hypopharyngeal SCC) cell lines re-expressing PKCßI and -ßII isoforms demonstrated IC50 of 1-5 ng/ml with TPA or PEP005. CONCLUSION: Re-expression of PKCß in head and neck SCC cell lines leads to cells one thousand-times more sensitive to the cytotoxic effects of phorbol or diterpene esters in culture. This highlights the importance of the isoform in tumor progression and presents the potential benefit of these compounds in malignancies expressing the protein, and in combination therapy.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Diterpenos/farmacologia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Proteína Quinase C beta/fisiologia , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/patologia , Sobrevivência Celular/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/enzimologia , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Isoenzimas/análise , Proteína Quinase C beta/análise , Carcinoma de Células Escamosas de Cabeça e Pescoço , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas
11.
J Neurosci ; 35(11): 4614-25, 2015 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-25788678

RESUMO

As one of the most effective and frequently used chemotherapeutic agents, paclitaxel produces peripheral neuropathy (paclitaxel-induced peripheral neuropathy or PIPN) that negatively affects chemotherapy and persists after cancer therapy. The mechanisms underlying this dose-limiting side effect remain to be fully elucidated. This study aimed to investigate the role of nociceptor protein kinase C (PKC) isoforms in PIPN. Employing multiple complementary approaches, we have identified a subset of PKC isoforms, namely ßII, δ, and ϵ, were activated by paclitaxel in the isolated primary afferent sensory neurons. Persistent activation of PKCßII, PKCδ, and PKCϵ was also observed in the dorsal root ganglion neurons after chronic treatment with paclitaxel in a mouse model of PIPN. Isoform-selective inhibitors of PKCßII, PKCδ, and PKCϵ given intrathecally dose-dependently attenuated paclitaxel-induced mechanical allodynia and heat hyperalgesia. Surprisingly, spinal inhibition of PKCßII and PKCδ, but not PKCϵ, blocked the spontaneous pain induced by paclitaxel. These data suggest that a subset of nociceptor PKC isoforms differentially contribute to spontaneous and evoked pain in PIPN, although it is not clear whether PKCϵ in other regions regulates spontaneous pain in PIPN. The findings can potentially offer new selective targets for pharmacological intervention of PIPN.


Assuntos
Nociceptores/fisiologia , Paclitaxel/toxicidade , Dor/enzimologia , Proteína Quinase C beta/fisiologia , Proteína Quinase C-delta/fisiologia , Proteína Quinase C-épsilon/fisiologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Nociceptores/efeitos dos fármacos , Dor/induzido quimicamente , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/fisiologia , Proteína Quinase C beta/antagonistas & inibidores , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-épsilon/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley
12.
Am J Physiol Lung Cell Mol Physiol ; 308(8): L827-36, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25659900

RESUMO

Pulmonary hypertension (PH) eventually leads to right ventricular (RV) fibrosis and dysfunction that is associated with increased morbidity and mortality. Although angiotensin II plays an important role in RV remodeling associated with hypoxic PH, the molecular mechanisms underlying RV fibrosis in PH largely remain unresolved. We hypothesized that PKC-p38 signaling is involved in RV collagen accumulation in PH and in response to angiotensin II stimulation. Adult male Sprague-Dawley rats were exposed to 3 wk of normoxia or hypoxia (10% FiO2 ) as a model of PH. Hypoxic rats developed RV hypertrophy and fibrosis associated with an increase in PKC ßII and δ protein expression and p38 dephosphorylation in freshly isolated RV cardiac fibroblasts. Further mechanistic studies were performed in cultured primary cardiac fibroblasts stimulated with angiotensin II, a key activator of ventricular fibrosis in PH. Angiotensin II induced a reduction in p38 phosphorylation that was attenuated following chemical inhibition of PKC ßII and δ. Molecular and chemical inhibition of PKC ßII and δ abrogated angiotensin II-induced cardiac fibroblast proliferation and collagen deposition in vitro. The effects of PKC inhibition on proliferation and fibrosis were reversed by chemical inhibition of p38. Conversely, constitutive activation of p38 attenuated angiotensin II-induced increase of cardiac fibroblast proliferation and collagen accumulation. PKC ßII- and δ-dependent inactivation of p38 regulates cardiac fibroblast proliferation and collagen deposition in response to angiotensin II, which suggests that the PKC-p38 signaling in cardiac fibroblasts may be involved and important in the pathophysiology of RV fibrosis in PH.


Assuntos
Angiotensina II/fisiologia , Hipertensão Pulmonar/enzimologia , Hipertrofia Ventricular Direita/enzimologia , Proteína Quinase C beta/fisiologia , Proteína Quinase C-delta/fisiologia , Animais , Hipóxia Celular , Proliferação de Células , Células Cultivadas , Colágeno/metabolismo , Ativação Enzimática , Fibroblastos/enzimologia , Fibrose , Ventrículos do Coração/patologia , Hipertensão Pulmonar/complicações , Masculino , Ratos Sprague-Dawley , Disfunção Ventricular Direita/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Naunyn Schmiedebergs Arch Pharmacol ; 387(5): 469-76, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24682423

RESUMO

The ultrarapidly activating delayed rectifier current, I(Kur), is a main determinant of atrial repolarization in humans. I(Kur) and the underlying ion channel complex Kv1.5/Kvß1.2 are negatively regulated by protein kinase C. However, the exact mode of action is only incompletely understood. We therefore analyzed isoenzyme-specific regulation of the Kv1.5/Kvß1.2 ion channel complex by PKC. Cloned ion channel subunits were heterologously expressed in Xenopus oocytes, and measurements were performed using the double-electrode voltage-clamp technique. Activation of PKC with phorbol 12-myristate 13-acetate (PMA) resulted in a strong reduction of Kv1.5/Kvß1.2 current. This effect could be prevented using the PKC inhibitor staurosporine. Using the bisindolylmaleimide Ro-31-8220 as an inhibitor and ingenol as an activator of the conventional PKC isoforms, we were able to show that the Kv1.5/Kvß1.2 ion channel complex is mainly regulated by conventional isoforms. Whereas pharmacological inhibition of PKCα with HBDDE did not attenuate the PMA-induced effect, current reduction could be prevented using inhibitors of PKCß. Here, we show the isoform ßII plays a central role in the PKC-dependent regulation of Kv1.5/Kvß1.2 channels. These results add to the current understanding of isoenzyme-selective regulation of cardiac ion channels by protein kinases.


Assuntos
Canal de Potássio Kv1.2/fisiologia , Canal de Potássio Kv1.5/fisiologia , Miocárdio/metabolismo , Proteína Quinase C beta/fisiologia , Animais , Humanos , Isoenzimas/fisiologia , Ligação Proteica , Acetato de Tetradecanoilforbol/farmacologia , Xenopus
14.
Mol Biol Cell ; 25(9): 1446-57, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24600048

RESUMO

Chemotaxis is a process by which cells polarize and move up a chemical gradient through the spatiotemporal regulation of actin assembly and actomyosin contractility, which ultimately control front protrusions and back retractions. We previously demonstrated that in neutrophils, mammalian target of rapamycin complex 2 (mTORC2) is required for chemoattractant-mediated activation of adenylyl cyclase 9 (AC9), which converts ATP into cAMP and regulates back contraction through MyoII phosphorylation. Here we study the mechanism by which mTORC2 regulates neutrophil chemotaxis and AC9 activity. We show that inhibition of protein kinase CßII (PKCßII) by CPG53353 or short hairpin RNA knockdown severely inhibits chemoattractant-induced cAMP synthesis and chemotaxis in neutrophils. Remarkably, PKCßII-inhibited cells exhibit specific and severe tail retraction defects. In response to chemoattractant stimulation, phosphorylated PKCßII, but not PKCα, is transiently translocated to the plasma membrane, where it phosphorylates and activates AC9. mTORC2-mediated PKCßII phosphorylation on its turn motif, but not its hydrophobic motif, is required for membrane translocation of PKCßII. Inhibition of mTORC2 activity by Rictor knockdown not only dramatically decreases PKCßII activity, but it also strongly inhibits membrane translocation of PKCßII. Together our findings show that PKCßII is specifically required for mTORC2-dependent AC9 activation and back retraction during neutrophil chemotaxis.


Assuntos
AMP Cíclico/biossíntese , Complexos Multiproteicos/metabolismo , Neutrófilos/enzimologia , Proteína Quinase C beta/fisiologia , Receptores de Formil Peptídeo/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Adenilil Ciclases/metabolismo , Membrana Celular/enzimologia , Quimiotaxia de Leucócito , Ativação Enzimática , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Alvo Mecanístico do Complexo 2 de Rapamicina , Miosina Tipo II , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/fisiologia , Fosforilação , Ftalimidas/farmacologia , Proteína Quinase C beta/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Transporte Proteico , Sistemas do Segundo Mensageiro
15.
Acta Pharm ; 64(1): 105-15, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24670355

RESUMO

C57BL/6 mice with dilated cardiomyopathy (DCM) were randomly divided to receive placebo or pitavastatin at a dose of 1 or 3 mg kg-1d-1. After 8 weeks treatment, mice with dilated cardiomyopathy developed serious cardiac dysfunction characterized by significantly enhanced left ventricular end-diastolic diameter (LVIDd), decreased left ventricular ejection fraction (LVEF) as well as left ventricular short axis fractional shortening (LVFS), accompanied with enlarged cardiomyocytes, and increased plasma levels of N-terminal pro-B type natriuretic peptide (NT-proBNP) and plasma angiotensin II (AngII) concentration. Moreover, myocardium sarcoplasmic reticulum Ca2+ pump (SERCA-2) activity was decreased. The ratio of phosphorylated phospholamban (PLB) to total PLB decreased significantly with the down-regulation of SERCA- -2a and ryanodine receptor (RyR2) expression. Pitavastatin was found to ameliorate the cardiac dysfunction in mice with dilated cardiomyopathy by reversing the changes in the ratios of phosphorylated PLB to total PLB, SERCA-2a and RyR2 via reducing the plasma AngII concentration and the expressions of myocardium angiotensin II type 1 receptor (AT1R) and protein kinase C (PKC)b2. The possible underlying mechanism might be the regulation of myocardial AT1R-PKCb2-Ca2+ handling proteins.


Assuntos
Cálcio/metabolismo , Cardiomiopatia Dilatada/tratamento farmacológico , Cardiomiopatia Dilatada/metabolismo , Proteínas de Membrana/fisiologia , Miocárdio/metabolismo , Quinolinas/uso terapêutico , Angiotensina II/fisiologia , Animais , Cardiomiopatia Dilatada/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeo Natriurético Encefálico/fisiologia , Fragmentos de Peptídeos/fisiologia , Proteína Quinase C beta/fisiologia , Quinolinas/farmacologia , Distribuição Aleatória , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/fisiologia
16.
Pigment Cell Melanoma Res ; 27(3): 387-97, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24472179

RESUMO

Syndecan-2, a transmembrane heparan sulfate proteoglycan that is highly expressed in melanoma cells, regulates melanoma cell functions (e.g. migration). Since melanoma is a malignant tumor of melanocytes, which largely function to synthesize melanin, we investigated the possible involvement of syndecan-2 in melanogenesis. Syndecan-2 expression was increased in human skin melanoma tissues compared with normal skin. In both mouse and human melanoma cells, siRNA-mediated knockdown of syndecan-2 was associated with reduced melanin synthesis, whereas overexpression of syndecan-2 increased melanin synthesis. Similar effects were also detected in human primary epidermal melanocytes. Syndecan-2 expression did not affect the expression of tyrosinase, a key enzyme in melanin synthesis, but instead enhanced the enzymatic activity of tyrosinase by increasing the membrane and melanosome localization of its regulator, protein kinase CßII. Furthermore, UVB caused increased syndecan-2 expression, and this up-regulation of syndecan-2 was required for UVB-induced melanin synthesis. Taken together, these data suggest that syndecan-2 regulates melanin synthesis and could be a potential therapeutic target for treating melanin-associated diseases.


Assuntos
Melaninas/biossíntese , Melanócitos/metabolismo , Melanoma/metabolismo , Monofenol Mono-Oxigenase/metabolismo , Proteína Quinase C beta/fisiologia , Sindecana-2/fisiologia , Animais , Linhagem Celular Tumoral , Membrana Celular/enzimologia , Ativação Enzimática , Células Epidérmicas , Epiderme/metabolismo , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Melanócitos/efeitos da radiação , Melanoma/patologia , Melanossomas/enzimologia , Camundongos , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Sindecana-2/antagonistas & inibidores , Sindecana-2/genética , Raios Ultravioleta , Regulação para Cima/efeitos da radiação
17.
Biochem Pharmacol ; 88(2): 139-49, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24440741

RESUMO

Atherosclerosis is a pathologic condition caused by chronic inflammation in response to lipid deposition in the arterial wall. There are many known contributing factors such as long-term abnormal glucose levels, smoking, hypertension, and hyperlipidemia. Under the influence of such factors, immune and non-immune effectors cells are activated and participate during the progression of atherosclerosis. Protein kinase C (PKC) family isoforms are key players in the signal transduction pathways of cellular activation and have been associated with several aspects of the atherosclerotic vascular disease. This review article summarizes the current knowledge of PKC isoforms functions during atherogenesis, and addresses differential roles and disputable observations of PKC isoforms. Among PKC isoforms, both PKCß and PKCδ are the most attractive and potential therapeutic targets. This commentary discusses in detail the outcomes and current status of clinical trials on PKCß and PKCδ inhibitors in atherosclerosis-associated disorders like diabetes and myocardial infarction. The risk and benefit of these inhibitors for clinical purposes will be also discussed. This review summarizes what is already being done and what else needs to be done in further targeting PKC isoforms, especially PKCß and PKCδ, for therapy of atherosclerosis and atherosclerosis-associated vasculopathies in the future.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Aterosclerose/enzimologia , Mediadores da Inflamação/fisiologia , Proteína Quinase C beta/fisiologia , Proteína Quinase C-delta/fisiologia , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Aterosclerose/tratamento farmacológico , Ensaios Clínicos como Assunto/métodos , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/química , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Isoenzimas/fisiologia , Proteína Quinase C beta/antagonistas & inibidores , Proteína Quinase C beta/química , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/química , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
18.
Pigment Cell Melanoma Res ; 27(3): 418-30, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24406113

RESUMO

A large-scale RNAi screen was performed for eight different melanoma cell lines using a pooled whole-genome lentiviral shRNA library. shRNAs affecting proliferation of transduced melanoma cells were negatively selected during 10 days of culture. Overall, 617 shRNAs were identified by microarray hybridization. Pathway analyses identified mitogen-activated protein kinase (MAPK) pathway members such as ERK1/2, JNK1/2 and MAP3K7 and protein kinase C ß (PKCß) as candidate genes. Knockdown of PKCß most consistently reduced cellular proliferation, colony formation and migratory capacity of melanoma cells and was selected for further validation. PKCß showed enhanced expression in human primary melanomas and distant metastases as compared with benign melanocytic nevi. Moreover, treatment of melanoma cells with PKCß-specific inhibitor enzastaurin reduced melanoma cell growth but had only small effects on benign fibroblasts. Finally, PKCß-shRNA significantly reduced lung colonization capacity of stably transduced melanoma cells in mice. Taken together, this study identified new candidate genes for melanoma cell growth and proliferation. PKCß seems to play an important role in these processes and might serve as a new target for the treatment of metastatic melanoma.


Assuntos
Sistema de Sinalização das MAP Quinases/fisiologia , Melanoma/patologia , Proteínas de Neoplasias/fisiologia , Proteína Quinase C beta/fisiologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Indução Enzimática , Regulação Neoplásica da Expressão Gênica , Biblioteca Genômica , Humanos , Indóis/farmacologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/enzimologia , Melanoma/prevenção & controle , Melanoma/secundário , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Nevo Pigmentado/enzimologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Quinase C beta/antagonistas & inibidores , Proteína Quinase C beta/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/genética , Proteínas Quinases/fisiologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/fisiologia , Neoplasias Cutâneas/enzimologia , Transdução Genética , Ensaio Tumoral de Célula-Tronco , Regulação para Cima
19.
Can J Physiol Pharmacol ; 91(11): 901-12, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24117257

RESUMO

This study aimed at investigating the efficacy of Kalpaamruthaa (KA) on cardiovascular damage (CVD) associated with type 2 diabetes mellitus in experimental rats by reducing oxidative stress and the modulation of the protein kinase C-ß (PKC-ß)/Akt signaling pathway. CVD-induced rats were treated with KA (200 mg·(kg body mass)(-1)·(day)(-1)) orally for 4 weeks. KA effectively reduced insulin resistance with alterations in blood glucose, hemoglobin, and glycosylated hemoglobin in CVD-induced rats. Elevated levels of lipids in CVD-induced rats were decreased upon KA administration. In CVD-induced rats the levels of lipoproteins were returned to normal by KA treatment. KA effectively reduced the lipid peroxidative product and protein carbonyl content in liver of CVD-induced rats. KA increased the activities and (or) levels of enzymatic and nonenzymatic antioxidants in liver of CVD-induced rats. KA treatment reduced the fatty inclusion and mast cell infiltration in liver of CVD-induced rats. Further, treatment with KA reduced the chromatin condensation and marginization in myocardium of CVD-induced rats. KA alters insulin signaling by decreasing PKC-ß and increasing p-Akt and GLUT4 expressions in heart of CVD-induced rats. The above findings suggest that KA renders protection against CVD induced by type 2 diabetes mellitus by augmenting the cellular antioxidant defense capacity and modulating PKC-ß and the p-Akt signaling pathway.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Proteína Quinase C beta/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Animais , Glicemia/metabolismo , Western Blotting , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatias Diabéticas/patologia , Transportador de Glucose Tipo 4/metabolismo , Imuno-Histoquímica , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipídeos/sangue , Lipoproteínas/sangue , Masculino , Mastócitos/efeitos dos fármacos , Microscopia Eletrônica de Transmissão , Miocárdio/patologia , Carbonilação Proteica/efeitos dos fármacos , Proteína Quinase C beta/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...