Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Mol Cell Cardiol ; 127: 125-133, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30550751

RESUMO

Huntington's disease (HD) is a fatal hereditary neurodegenerative disorder, best known for its clinical triad of progressive motor impairment, cognitive deficits and psychiatric disturbances, is caused by CAG-repeat expansion in exon 1 of Huntingtin (HTT). However, in addition to the neurological disease, mutant HTT (mHTT), which is ubiquitously expressed in all tissues, impairs other organ systems. Not surprisingly, cardiovascular dysautonomia as well as the deterioration of circadian rhythms are among the earliest detectable pathophysiological changes in individuals with HD. Mitochondrial dysfunction in the brain and skeletal muscle in HD has been well documented, as the disease progresses. However, not much is known about mitochondrial abnormalities in the heart. In this study, we describe a role for Drp1/Fis1-mediated excessive mitochondrial fission and dysfunction, associated with lysosomal dysfunction in H9C2 expressing long polyglutamine repeat (Q73) and in human iPSC-derived cardiomyocytes transfected with Q77. Expression of long polyglutamine repeat led to reduced ATP production and mitochondrial fragmentation. We observed an increased accumulation of damaged mitochondria in the lysosome that was coupled with lysosomal dysfunction. Importantly, reducing Drp1/Fis1-mediated mitochondrial damage significantly improved mitochondrial function and cell survival. Finally, reducing Fis1-mediated Drp1 recruitment to the mitochondria, using the selective inhibitor of this interaction, P110, improved mitochondrial structure in the cardiac tissue of R6/2 mice. We suggest that drugs focusing on the central nervous system will not address mitochondrial function across all organs, and therefore will not be a sufficient strategy to treat or slow down HD disease progression.


Assuntos
Dinaminas/metabolismo , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Lisossomos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Metabolismo Energético , GTP Fosfo-Hidrolases/farmacologia , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lisossomos/ultraestrutura , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/ultraestrutura , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos/metabolismo , Expansão das Repetições de Trinucleotídeos
5.
Oncogene ; 31(20): 2593-600, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21996750

RESUMO

Protein kinase C (PKC)ɛ, a member of the novel PKC family, has key roles in mitogenesis and survival in normal and cancer cells. PKCɛ is frequently overexpressed in epithelial cancers, particularly in lung cancer. Using a short-hairpin RNA approach, here we established that PKCɛ is required for non-small cell lung carcinoma (NSCLC) growth in vitro as well as tumor growth when inoculated into athymic mice. Moreover, sustained delivery of a PKCɛ-selective inhibitor peptide, ɛV1-2, reduced xenograft growth in mice. Both RNA interference depletion and pharmacological inhibition of PKCɛ caused a marked elevation in the number of apoptotic cells in NSCLC tumors. PKCɛ-depleted NSCLC cells show elevated expression of pro-apoptotic proteins of the Bcl-2 family, caspase recruitment domain-containing proteins and tumor necrosis factor ligands/receptor superfamily members. Moreover, a Gene Set Enrichment Analysis revealed that a vast majority of the genes changed in PKCɛ-depleted cells were also deregulated in human NSCLC. Our results strongly suggest that PKCɛ is required for NSCLC cell survival and maintenance of NSCLC tumor growth. Therefore, PKCɛ may represent an attractive therapeutic target for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/enzimologia , Neoplasias Pulmonares/enzimologia , Proteína Quinase C-épsilon/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Genes bcl-2 , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fragmentos de Peptídeos/farmacologia , Proteína Quinase C-épsilon/antagonistas & inibidores , Proteína Quinase C-épsilon/genética , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Receptores do Fator de Necrose Tumoral/metabolismo
6.
PLoS ONE ; 7(12): p.12, 2012.
Artigo em Inglês | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib10819
7.
Oncogene ; 30(3): 323-33, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-20856202

RESUMO

Metastasis is the major reason for breast cancer-related deaths. Although there is a host of indirect evidence for a role of protein kinase C (PKC) α in primary breast cancer growth, its role in the molecular pathways leading to metastasis has not been studied comprehensively. By treating mice with αV5-3, a novel peptide inhibitor selective for PKCα, we were able to determine how PKCα regulates metastasis of mammary cancer cells using a syngeneic and orthotopic model. The primary tumor growth was not affected by αV5-3 treatment. However, the mortality rate was reduced and metastasis in the lung decreased by more than 90% in the αV5-3-treated mice relative to the control-treated mice. αV5-3 treatment reduced intravasation by reducing matrix metalloproteinase-9 activities. αV5-3 treatment also reduced lung seeding of tumor cells and decreased cell migration, effects that were accompanied by a reduction in nuclear factor kappa B activity and cell surface levels of the CXCL12 receptor, CXCR4. αV5-3 treatment caused no apparent toxicity in non-tumor-bearing naïve mice. Rather, inhibiting PKCα protected against liver damage and increased the number of immune cells in tumor-bearing mice. Importantly, αV5-3 showed superior efficacy relative to anti-CXCR4 antibody in reducing metastasis in vivo. Together, these data show that pharmacological inhibition of PKCα effectively reduces mammary cancer metastasis by targeting intravasation and lung seeding steps in the metastatic process and suggest that PKCα-specific inhibitors, such as αV5-3, can be used to study the mechanistic roles of PKCα specifically and may provide a safe and effective treatment for the prevention of lung metastasis of breast cancer patients.


Assuntos
Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Metástase Neoplásica , Inoculação de Neoplasia , Proteína Quinase C-alfa/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase C-alfa/genética , Proteína Quinase C-alfa/metabolismo , RNA Interferente Pequeno , Receptores CXCR4/metabolismo
8.
Transplant Proc ; 40(2): 375-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18374073

RESUMO

Strategies inhibiting cell death signaling pathways may enhance the availability of islet transplantation for patients with type 1 diabetes mellitus. The epsilon isoform of protein kinase C (PKC epsilon) has been shown to have an anti-apoptotic effect in many cell types. The present study investigated whether activation of PKC epsilon may improve the yield of functional islet cells for transplantation. Islet cells were isolated from wild-type BALB/c mice preconditioned with either a PKC epsilon activator (psi epsilon RACK) or a TAT carrier control peptide and further treated with the same agents during isolation and in vitro for either 0, 1, 16, or 40 hours. Islet cells were assessed at each time point for viability, apoptosis, and function. psi epsilon RACK-treated islets showed significantly decreased islet cell death up to 40 hours after isolation compared with TAT-treated control islets. Beta-cell function in response to high glucose challenge remained unchanged.


Assuntos
Sobrevivência Celular/fisiologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/enzimologia , Proteína Quinase C-épsilon/metabolismo , Animais , Técnicas de Cultura de Células , Separação Celular/métodos , Diabetes Mellitus Tipo 1/cirurgia , Ativação Enzimática , Humanos , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Coleta de Tecidos e Órgãos/métodos
9.
Biochem Soc Trans ; 35(Pt 5): 1021-6, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17956268

RESUMO

PKC (protein kinase C) isoenzymes are related protein kinases, involved in many signalling events in normal state and in disease. Basic research into identifying the molecular basis of PKC selectivity led to simple strategies to identify selective competitive inhibitor peptides and allosteric agonist peptides of individual PKC isoenzymes. The strategies and rationale used to identify these peptide regulators of protein-protein interaction may be applicable to other signalling events. Importantly, the PKC-regulating peptides proved to be useful pharmacological tools and may serve as drugs or drug leads for a variety of human diseases.


Assuntos
Ativadores de Enzimas/farmacologia , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Sequência de Aminoácidos , Humanos , Isoenzimas/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Transdução de Sinais , Frações Subcelulares/enzimologia
10.
Biochem Soc Trans ; 35(Pt 5): 1040-2, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17956273

RESUMO

Reperfusion of ischaemic cardiac tissue is associated with increased apoptosis and oncosis, resulting in diminished heart function. Short bouts of ischaemia before the prolonged ischaemic event (ischaemic preconditioning) protect the heart from injury mediated by reperfusion. The PKC (protein kinase C) family of serine/threonine kinases are involved in many different signalling processes. Two calcium-insensitive isoforms of the novel PKC subfamily, PKCdelta and epsilon, play opposing roles in ischaemia/reperfusion injury. Activation of PKCdelta during reperfusion induces cell death through the regulation of mitochondrial function and induction of apoptosis and oncosis. In contrast, activation of PKCepsilon before ischaemia protects mitochondrial function and diminishes apoptosis and oncosis. How can two highly homologous PKC isoenzymes play such opposing roles through the regulation of mitochondrial function? This review will highlight what is known about PKCdelta and epsilon function during ischaemia/reperfusion injury and will suggest a novel regulatory pathway which determines the fate of the cell following ischaemic stress.


Assuntos
Isoenzimas/metabolismo , Traumatismo por Reperfusão Miocárdica/enzimologia , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-épsilon/metabolismo , Hidrólise , Complexo de Endopeptidases do Proteassoma/metabolismo
11.
Biochem Soc Trans ; 35(Pt 5): 1052-4, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17956277

RESUMO

Mitochondria mediate diverse cellular functions including energy generation and ROS (reactive oxygen species) production and contribute to signal transduction. Mitochondria are also key regulators of cell viability and play a central role in necrotic and apoptotic cell death pathways induced by cardiac ischaemia/reperfusion injury. PKC (protein kinase C) epsilon plays a critical role in cardioprotective signalling pathways that protect the heart from ischaemia/reperfusion. Emerging evidence suggests that the cardioprotective target of PKCepsilon resides at the mitochondria. Proposed mitochondrial targets of PKCepsilon include mitoK(ATP) (mitochondrial ATP-sensitive K(+) channel), components of the MPTP (mitochondrial permeability transition pore) and components of the electron transport chain. This review highlights mitochondrial targets of PKCepsilon and their possible role in cardioprotective signalling in the setting of ischaemia/reperfusion injury.


Assuntos
Mitocôndrias Cardíacas/enzimologia , Isquemia Miocárdica/prevenção & controle , Proteína Quinase C-épsilon/metabolismo , Isquemia Miocárdica/enzimologia , Transdução de Sinais
12.
J Mol Cell Cardiol ; 39(2): 203-11, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15990110

RESUMO

Previous studies have demonstrated that acute ethanol exposure induces activation of delta protein kinase C (deltaPKC) and epsilonPKC, and mimics ischemic preconditioning via epsilonPKC activation. However, the role of deltaPKC isozyme in ischemia and reperfusion is still controversial. Here, we investigated the role of deltaPKC in ethanol-induced cardioprotection using a selective deltaPKC activator (psideltaRACK), or inhibitor (deltaV1-1), and a selective epsilonPKC inhibitor (epsilonV1-2) in isolated mouse hearts. Mice were injected intraperitoneally or by gavage with ethanol, regulators of delta and epsilonPKC or an adenosine A1 receptor blocker (DPCPX). Isolated perfused mouse hearts were subjected to a 30-min global ischemia and a 120-min reperfusion, ex vivo. Injection of 0.5 g/kg ethanol 1 h, but not 10 min, before ischemia reduced infarct size and CPK release. Pretreatment with epsilonV1-2 abolished this ethanol-induced cardioprotection. Pretreatment with deltaV1-1 induced cardioprotection when injected with ethanol (0.5 g/kg) 10 min before ischemia, but deltaV1-1 partly inhibited ethanol-induced cardioprotection when injected with ethanol 1-h before the onset of ischemia. psideltaRACK injection 1 h, but not 10 min, before ischemia induced cardioprotection and translocation of epsilonPKC from the cytosol to the particulate fraction. Pretreatment with DPCPX or epsilonV1-2 inhibited psideltaRACK-induced cardioprotection and translocation of epsilonPKC. Therefore, activation of deltaPKC-induced by ethanol or by the deltaPKC activator is cardioprotective, provided that sufficient time passes to allow deltaPKC-induced activation of epsilonPKC, an A1 adenosine receptor-dependent process.


Assuntos
Etanol/farmacologia , Isquemia Miocárdica/prevenção & controle , Animais , Ativação Enzimática , Ativadores de Enzimas/metabolismo , Ativadores de Enzimas/farmacologia , Isoenzimas/metabolismo , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Transporte Proteico/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Frações Subcelulares/metabolismo
13.
Am J Physiol Cell Physiol ; 281(5): C1477-86, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11600410

RESUMO

Investigation of the role of individual protein kinase C (PKC) isozymes in the regulation of Na(+) channels has been largely limited by the lack of isozyme-selective modulators. Here we used a novel peptide-specific activator (epsilonV1-7) of epsilonPKC and other peptide isozyme-specific inhibitors in addition to the general PKC activator phorbol 12-myristate 13-acetate (PMA) to dissect the role of individual PKCs in the regulation of the human cardiac Na(+) channel hH1, heterologously expressed in Xenopus oocytes. Peptides were injected individually or in combination into the oocyte. Whole cell Na(+) current (I(Na)) was recorded using two-electrode voltage clamp. epsilonV1-7 (100 nM) and PMA (100 nM) inhibited I(Na) by 31 +/- 5% and 44 +/- 8% (at -20 mV), respectively. These effects were not seen with the scrambled peptide for epsilonV1-7 (100 nM) or the PMA analog 4alpha-phorbol 12,13-didecanoate (100 nM). However, epsilonV1-7- and PMA-induced I(Na) inhibition was abolished by epsilonV1-2, a peptide-specific antagonist of epsilonPKC. Furthermore, PMA-induced I(Na) inhibition was not altered by 100 nM peptide-specific inhibitors for alpha-, beta-, delta-, or etaPKC. PMA and epsilonV1-7 induced translocation of epsilonPKC from soluble to particulate fraction in Xenopus oocytes. This translocation was antagonized by epsilonV1-2. In native rat ventricular myocytes, PMA and epsilonV1-7 also inhibited I(Na); this inhibition was antagonized by epsilonV1-2. In conclusion, the results provide evidence for selective regulation of cardiac Na(+) channels by epsilonPKC isozyme.


Assuntos
Miocárdio/metabolismo , Proteína Quinase C/fisiologia , Canais de Sódio/fisiologia , Animais , Separação Celular , Clonagem Molecular , Técnicas In Vitro , Isoenzimas/fisiologia , Miocárdio/citologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Peptídeos/farmacologia , RNA Complementar/biossíntese , Ratos , Ratos Wistar , Agonistas de Canais de Sódio , Bloqueadores dos Canais de Sódio/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Xenopus
14.
Oncogene ; 20(44): 6339-47, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11607837

RESUMO

Spatial and temporal organization of signal transduction is essential in determining the speed and precision by which signaling events occur. Adaptor proteins are key to organizing signaling enzymes near their select substrates and away from others in order to optimize precision and speed of response. Here, we describe the role of adaptor proteins in determining the specific function of individual protein kinase C (PKC) isozymes. These isozyme-selective proteins were called collectively RACKs (receptors for activated C-kinase). The role of RACKs in PKC-mediated signaling was determined using isozyme-specific inhibitors and activators of the binding of each isozyme to its respective RACK. In addition to anchoring activated PKC isozymes, RACKs anchor other signaling enzymes. RACK1, the anchoring protein for activated betaIIPKC, binds for example, Src tyrosine kinase, integrin, and phosphodiesterase. RACK2, the epsilonPKC-specific RACK, is a coated-vesicle protein and thus is involved in vesicular release and cell-cell communication. Therefore, RACKs are not only adaptors for PKC, but also serve as adaptor proteins for several other signaling enzymes. Because at least some of the proteins that bind to RACKs, including PKC itself, regulate cell growth, modulating their interactions with RACKs may help elucidate signaling pathways leading to carcinogenesis and could result in the identification of novel therapeutic targets.


Assuntos
Proteína Quinase C/metabolismo , Transdução de Sinais , Animais , Humanos , Isoenzimas/metabolismo , Modelos Biológicos , Neoplasias/metabolismo , Neoplasias/prevenção & controle , Diester Fosfórico Hidrolases/metabolismo , Ligação Proteica , Proteínas Tirosina Quinases/metabolismo
15.
Proc Natl Acad Sci U S A ; 98(20): 11114-9, 2001 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-11553773

RESUMO

Conflicting roles for protein kinase C (PKC) isozymes in cardiac disease have been reported. Here, deltaPKC-selective activator and inhibitor peptides were designed rationally, based on molecular modeling and structural homology analyses. Together with previously identified activator and inhibitor peptides of epsilonPKC, deltaPKC peptides were used to identify cardiac functions of these isozymes. In isolated cardiomyocytes, perfused hearts, and transgenic mice, deltaPKC and epsilonPKC had opposing actions on protection from ischemia-induced damage. Specifically, activation of epsilonPKC caused cardioprotection whereas activation of deltaPKC increased damage induced by ischemia in vitro and in vivo. In contrast, deltaPKC and epsilonPKC caused identical nonpathological cardiac hypertrophy; activation of either isozyme caused nonpathological hypertrophy of the heart. These results demonstrate that two related PKC isozymes have both parallel and opposing effects in the heart, indicating the danger in the use of therapeutics with nonselective isozyme inhibitors and activators. Moreover, reduction in cardiac damage caused by ischemia by perfusion of selective regulator peptides of PKC through the coronary arteries constitutes a major step toward developing a therapeutic agent for acute cardiac ischemia.


Assuntos
Cardiomegalia/fisiopatologia , Coração/efeitos dos fármacos , Isoenzimas/metabolismo , Contração Miocárdica/fisiologia , Isquemia Miocárdica/fisiopatologia , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Proteína Quinase C/metabolismo , Sequência de Aminoácidos , Animais , Células Cultivadas , Ativação Enzimática , Coração/fisiologia , Coração/fisiopatologia , Hemodinâmica/efeitos dos fármacos , Hemodinâmica/fisiologia , Técnicas In Vitro , Isoenzimas/química , Masculino , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Dados de Sequência Molecular , Contração Miocárdica/efeitos dos fármacos , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/química , Peptídeos/síntese química , Peptídeos/química , Conformação Proteica , Proteína Quinase C/química , Proteína Quinase C-delta , Proteína Quinase C-épsilon , Ratos , Ratos Wistar , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
16.
J Mol Cell Cardiol ; 33(7): 1301-7, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11437536

RESUMO

Although protein kinase C (PKC) was identified more than 20 years ago, and is involved in a wide variety of essential cellular processes, assigning specific roles to each PKC isozyme has proved difficult. Results over the last few years have suggested that much of the specificity of activated PKC isozymes is attributed to their subcellular localization bringing them into close proximity to a subset of substrates. Our laboratory has taken advantage of the importance of PKC localization and studied the way in which PKC isozymes are anchored. We have identified PKC anchoring proteins (RACKs or Receptors for Activated C Kinase) and used information about interaction sites between PKC isozymes and their respective RACKs to design peptides which modulate translocation of specific PKC isozymes to the functional site. These isozyme-specific peptides can be delivered into isolated or cultured cells or expressed in transgenic mice to determine the role of specific PKC isozymes in particular functions. Here we will describe the isozymes-specific peptide activators and inhibitors that we have developed and the specific functions of each isozyme in cardiac ventricular tissue.


Assuntos
Coração/fisiologia , Isoenzimas/química , Miocárdio/metabolismo , Peptídeos/metabolismo , Proteína Quinase C/química , Proteína Quinase C/fisiologia , Animais , Ácido Araquidônico/metabolismo , Ácido Araquidônico/farmacologia , Etanol/metabolismo , Etanol/farmacologia , Coração/efeitos dos fármacos , Precondicionamento Isquêmico , Isoenzimas/metabolismo , Peptídeos/farmacologia , Substâncias Protetoras/metabolismo , Substâncias Protetoras/farmacologia , Proteína Quinase C/antagonistas & inibidores , Transporte Proteico/efeitos dos fármacos , Receptores de Quinase C Ativada
18.
J Biol Chem ; 276(32): 29644-50, 2001 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-11387319

RESUMO

Identification of selective anchoring proteins responsible for specialized localization of specific signaling proteins has led to the identification of new inhibitors of signal transduction, inhibitors of anchoring protein-ligand interactions. RACK1, the first receptor for activated C kinase identified in our lab, is a selective anchoring protein for betaII protein kinase C (betaIIPKC). We previously found that at least part of the RACK1-binding site resides in the C2 domain of betaIIPKC (Ron, D., Luo, J., and Mochly-Rosen, D. (1995) J. Biol. Chem. 270, 24180-24187). Here we show that the V5 domain also contains part of the RACK1-binding site in betaIIPKC. In neonatal rat cardiac myocytes, the betaIIV5-3 peptide (amino acids 645-650 in betaIIPKC) selectively inhibited phorbol 12-myristate 13-acetate (PMA)-induced translocation of betaIIPKC and not betaIPKC. In addition, the betaIIV5-3 peptide inhibited cardiac myocyte hypertrophy in PMA-treated cells. Interestingly, betaIV5-3 (646-651 in betaIPKC), a selective translocation inhibitor of betaIPKC, also inhibited PMA-induced cardiac myocyte hypertrophy, demonstrating that both betaI- and betaIIPKC are essential for this cardiac function. Therefore, the betaIIV5 domain contains part of the RACK1-binding site in betaIIPKC; a peptide corresponding to this site is a selective inhibitor of betaIIPKC and, hence, enables the identification of betaIIPKC-selective functions.


Assuntos
Isoenzimas/química , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteína Quinase C/química , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Sítios de Ligação , Ligação Competitiva , Western Blotting , Células Cultivadas , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP , Glutationa Transferase/metabolismo , Microscopia de Fluorescência , Dados de Sequência Molecular , Miocárdio/citologia , Peptídeos/química , Fenilalanina/metabolismo , Ligação Proteica , Proteína Quinase C beta , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Receptores de Quinase C Ativada , Receptores de Superfície Celular , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia
19.
Cardiovasc Res ; 50(1): 65-74, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11282079

RESUMO

OBJECTIVES: Arachidonic acid is a second messenger which activates protein kinase C (PKC) and is released from the heart during ischaemic preconditioning. The purpose of this study was to examine the effect of arachidonic acid on activation of PKC in cardiac myocytes and the cellular consequences. METHODS: Neonatal rat cardiac myocytes were isolated and maintained in culture. Arachidonic acid-induced activation of PKC was examined by cell fractionation and western blot analysis. Contraction frequency was measured by visual inspection under a microscope. Ischaemia was simulated by subjecting cells to an atmosphere of lower than 0.5% oxygen in the absence of glucose and cell damage determined by release of cytosolic lactate dehydrogenase or direct cell viability assay. RESULTS: Arachidonic acid resulted in translocation of delta and epsilonPKC but not alpha, betaII, eta or zetaPKC isozymes, indicating activation of only delta and epsilonPKC. Arachidonic acid induced a dose-dependent decrease in spontaneous contraction rate of cardiac myocytes which was blocked by a selective peptide translocation inhibitor of epsilonPKC. Pretreatment with arachidonic acid partially protected cardiac myocytes against ischaemia. Down-regulation of PKC with 24 h 4beta-phorbol,12-myristate,13-acetate treatment, inhibition of PKC by chelerythrine and selective inhibition of epsilonPKC translocation all decreased the protective effect of arachidonic acid. Pretreatment with eicosapentaenoic acid or oleic acid also protected cardiac myocytes against ischaemia. CONCLUSIONS: These results demonstrate that arachidonic acid selectively activates delta and epsilonPKC in neonatal rat cardiac myocytes, leading to protection from ischaemia. We suggest this is a potential mechanism of PKC activation during PC. In addition, our results suggest that different classes of free fatty acid directly exert cardioprotection from ischaemic injury in cardiac myocytes.


Assuntos
Ácido Araquidônico/farmacologia , Precondicionamento Isquêmico Miocárdico , Isquemia Miocárdica/prevenção & controle , Miocárdio/enzimologia , Proteína Quinase C/metabolismo , Animais , Técnicas de Cultura de Células , Relação Dose-Resposta a Droga , Ativação Enzimática , Contração Miocárdica/efeitos dos fármacos , Isquemia Miocárdica/patologia , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/fisiologia , Ratos , Ratos Sprague-Dawley , Translocação Genética/efeitos dos fármacos
20.
J Mol Cell Cardiol ; 33(3): 581-5, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11181025

RESUMO

Low amounts of ethanol reduce cardiac damage induced by ischemia. The protection from ischemic damage by acute exposure to low amounts of ethanol in isolated myocytes and intact heart have been attributed to activation of protein kinase C (PKC). We previously found that two PKC isozymes, delta and xi, are activated by ethanol in several cell models. Here, we perfused isozyme-selective agonist and antagonist peptides that we have generated into intact heart to determine the role of these two isozymes in ethanol-induced protection from transient ischemia. Whereas xi PKC activation was required for ethanol-induced protection, delta PKC activation led to further damage. These data explain the conflicting reports on the role of acute exposure to ethanol in protection from cardiac ischemia. The clinical implications of these findings are also discussed.


Assuntos
Fármacos Cardiovasculares/farmacologia , Etanol/farmacologia , Traumatismos Cardíacos/prevenção & controle , Isoenzimas/fisiologia , Isquemia Miocárdica/patologia , Substâncias Protetoras/farmacologia , Proteína Quinase C/fisiologia , Animais , Ativação Enzimática , Isoenzimas/antagonistas & inibidores , Masculino , Camundongos , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-delta , Proteína Quinase C-épsilon
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...