Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Neurovasc Res ; 8(3): 200-9, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21675954

RESUMO

We demonstrated that the endocannabinoid 2-arachidonoylglycerol (2-AG) activated dose-dependently washed human platelets and increased intracellular calcium levels. Moreover 2-AG activated protein kinase C measured as p47pleckstrin phosphorylation. These parameters were prevented by the tromboxane A2 receptor antagonist SQ29548, by phospholipase C pathway (U73122) and protein kinase C (GF109203X) inhibitors. No effect on 2-AG-induced platelet activation and calcium elevation in the presence of inhibitors of fatty acid amide hydrolase or monoacylglycerol lipase was observed. In addition we have shown that 2-AG dose-dependently increased NO and cGMP levels. These effects were abolished by U73122, GF109203X, EGTA and the intracellular calcium chelator BAPTA/AM. Moreover, 2-AG enhanced eNOS activity through the phosphorylation of its positive regulatory residue ser1177 and by dephosphorylation of the negative one thr495. The eNOS ser1177 phosphorylation was inhibited by U73122 and GF109203X but it was unaffected by the PI3K/AKT pathway inhibitors LY294002 and MK2206. The dephosphorylation of thr495 was reversed by low concentrations of calyculin A. Taken together these data suggest that 2-AG behaves as a true platelet agonist stimulating PKC activation and calcium elevation. Likely 2-AG can modulate platelet activation by increasing NO levels through eNOS activation.


Assuntos
Ácidos Araquidônicos/metabolismo , Plaquetas/metabolismo , Moduladores de Receptores de Canabinoides/metabolismo , Glicerídeos/metabolismo , Ativação Plaquetária , Proteína Quinase C/metabolismo , Transdução de Sinais , Ácidos Araquidônicos/farmacologia , Moduladores de Receptores de Canabinoides/farmacologia , GMP Cíclico/metabolismo , Endocanabinoides , Ativação Enzimática/fisiologia , Glicerídeos/farmacologia , Humanos , Immunoblotting , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Transdução de Sinais/efeitos dos fármacos
2.
Thromb Res ; 124(6): e48-55, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19660790

RESUMO

INTRODUCTION: Retinal vein occlusion is a major cause of ocular morbidity. The precise mechanism leading to thrombosis in retinal vein occlusion has not yet been clearly elucidated. Several risk factors have been identified, including hypertension diabetes, history of cardiovascular disease, hypercholesterolemia, hyperhomocysteinaemia, increased ocular pressure and glaucoma. Although thrombus formation in the vein plays a significant role in the onset of retinal vein occlusion, the relationship between platelet aggregation and retinal vein occlusion remains to be clarified. MATERIALS AND METHODS: In the present study the platelet response to thrombin in a selected group of retinal vein occlusion patients was investigated. Retinal vein occlusion patients were compared to a group of healthy subjects matched for age, sex, clinical and metabolic characteristics. In resting and activated platelets of both groups of subjects total protein tyrosine phosphorylation, p38MAPK and cytosolic phospholipase A(2) phosphorylation, arachidonic acid release, intracellular calcium levels, thromboxane B(2) and superoxide anion formation were measured. RESULTS: Results show that platelets of patients were more responsive to thrombin than healthy subjects. In resting or in thrombin stimulated platelets of patients total protein tyrosine phosphorylation, p38MAPK and cytosolic phospholipase A(2) phosphorylation were increased. Also arachidonic acid release, thromboxane B(2) and superoxide anion formation were higher in patients than in healthy subjects. In addition intracellular calcium rise induced by thrombin was increased in patients. CONCLUSIONS: Altogether data suggest that platelet hyperaggregability inducing thrombus formation might be an important factor in the onset and/or development of retinal vein occlusion.


Assuntos
Plaquetas/metabolismo , Retina/metabolismo , Oclusão da Veia Retiniana/fisiopatologia , Vasos Retinianos/fisiologia , Trombina/metabolismo , Idoso , Ácido Araquidônico/metabolismo , Estudos de Casos e Controles , Citosol/enzimologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosfolipases A2/metabolismo , Oclusão da Veia Retiniana/etiologia , Superóxidos/metabolismo , Tromboxano B2/metabolismo , Resultado do Tratamento , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Biochim Biophys Acta ; 1791(11): 1084-92, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19615463

RESUMO

Arachidonic acid can act as a second messenger regulating many cellular processes among which is nitric oxide (NO) formation. The aim of the present study was to investigate the molecular mechanisms involved in the arachidonic acid effect on platelet NO level. Thus NO, cGMP and superoxide anion level, the phosphorylation status of nitric oxide synthase, the protein kinase C (PKC), and NADPH oxidase activation were measured. Arachidonic acid dose-dependently reduced NO and cGMP level. The thromboxane A(2) mimetic U46619 behaved in a similar way. The arachidonic acid or U46619 effect on NO concentration was abolished by the inhibitor of the thromboxane A(2) receptor SQ29548 and partially reversed by the PKC inhibitor GF109203X or by the phospholipase C pathway inhibitor U73122. Moreover, it was shown that arachidonic acid activated PKC and decreased nitric oxide synthase (eNOS) activities. The phosphorylation of the inhibiting eNOSthr495 residue mediated by PKC was increased by arachidonic acid, while no changes at the activating ser1177 residue were shown. Finally, arachidonic acid induced NADPH oxidase activation and superoxide anion formation. These effects were greatly reduced by GF109203X, U73122, and apocynin. Likely arachidonic acid reducing NO bioavailability through all these mechanisms could potentiate its platelet aggregating power.


Assuntos
Ácido Araquidônico/farmacologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Óxido Nítrico/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Plaquetas/enzimologia , Cálcio/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , GMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , NADPH Oxidases/metabolismo , Nitratos/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Nitritos/metabolismo , Fosforilação/efeitos dos fármacos , Fosfotreonina/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Proteína Quinase C/metabolismo , Transporte Proteico/efeitos dos fármacos , Superóxidos/metabolismo
4.
Nitric Oxide ; 20(2): 104-13, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19100855

RESUMO

Hyperhomocysteinaemia has been associated with increased risk of thrombosis and atherosclerosis. Homocysteine produces endothelial injury and stimulates platelet aggregation. Several molecular mechanisms related to these effects have been elucidated. The study aimed to deeply investigate the homocysteine effect on nitric oxide formation in human platelets. The homocysteine-induced changes on nitric oxide, cGMP, superoxide anion levels and nitrotyrosine formation were evaluated. The enzymatic activity and the phosphorylation status of endothelial nitric oxide synthase (eNOS) at thr495 and ser1177 residues were measured. The protein kinase C (PKC), assayed by immunofluorescence confocal microscopy technique and by phosphorylation of p47pleckstrin, and NADPH oxidase activation, tested by the translocation to membrane of the two cytosolic subunits p47(phox) and p67(phox), were assayed. Results show that homocysteine reduces platelet nitric oxide and cGMP levels. The inhibition of eNOS activity and the stimulation of NADPH oxidase primed by PKC appear to be involved. PKC stimulates the eNOS phosphorylation of the negative regulatory residue thr495 and the dephosphorylation of the positive regulatory site ser1177. GF109203X and U73122, PKC and phospholipase Cgamma2 pathway inhibitors, respectively, reverse this effect. Moreover, homocysteine stimulates superoxide anion elevation and NADPH oxidase activation. These effects are significantly decreased by GF109203X and U73122, suggesting the involvement of PKC in NADPH oxidase activation. Homocysteine induces formation of the peroxynitrite biomarker nitrotyrosine. Taken together these results suggest that the homocysteine-mediated responses leading to nitric oxide impairment are mainly coupled to PKC activation. Thus homocysteine stimulates platelet aggregation and decreases nitric oxide bioavailability.


Assuntos
Plaquetas/metabolismo , Homocisteína/metabolismo , Óxido Nítrico/sangue , Proteína Quinase C/sangue , Análise de Variância , Plaquetas/citologia , Citrulina/sangue , GMP Cíclico/sangue , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Homocisteína/farmacologia , Humanos , Microscopia de Fluorescência , NADPH Oxidases/sangue , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação/efeitos dos fármacos , Superóxidos/sangue , Tirosina/análogos & derivados , Tirosina/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...