Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Blood ; 123(8): e11-22, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24408324

RESUMEN

Up to 1% of the population have mild bleeding disorders, but these remain poorly characterized, particularly with regard to the roles of platelets. We have compared the usefulness of Optimul, a 96-well plate-based assay of 7 distinct pathways of platelet activation to characterize inherited platelet defects in comparison with light transmission aggregometry (LTA). Using Optimul and LTA, concentration-response curves were generated for arachidonic acid, ADP, collagen, epinephrine, Thrombin receptor activating-peptide, U46619, and ristocetin in samples from (1) healthy volunteers (n = 50), (2) healthy volunteers treated with antiplatelet agents in vitro (n = 10), and (3) patients with bleeding of unknown origin (n = 65). The assays gave concordant results in 82% of cases (κ = 0.62, P < .0001). Normal platelet function results were particularly predictive (sensitivity, 94%; negative predictive value, 91%), whereas a positive result was not always substantiated by LTA (specificity, 67%; positive predictive value, 77%). The Optimul assay was significantly more sensitive at characterizing defects in the thromboxane pathway, which presented with normal responses with LTA. The Optimul assay is sensitive to mild platelet defects, could be used as a rapid screening assay in patients presenting with bleeding symptoms, and detects changes in platelet function more readily than LTA. This trial was registered at www.isrctn.org as #ISRCTN 77951167.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/diagnóstico , Monitoreo de Drogas/métodos , Hemorragia/diagnóstico , Ensayos Analíticos de Alto Rendimiento/métodos , Activación Plaquetaria/fisiología , Inhibidores de Agregación Plaquetaria/farmacología , Adulto , Trastornos de las Plaquetas Sanguíneas/sangre , Trastornos de las Plaquetas Sanguíneas/genética , Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Femenino , Estudios de Asociación Genética , Voluntarios Sanos , Hemorragia/sangre , Hemorragia/fisiopatología , Humanos , Masculino , Activación Plaquetaria/efectos de los fármacos , Valor Predictivo de las Pruebas , Receptores de Tromboxano A2 y Prostaglandina H2/genética , Sensibilidad y Especificidad , Adulto Joven
2.
Br J Clin Pharmacol ; 81(4): 621-33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26561399

RESUMEN

AIMS: In vivo platelet function is a product of intrinsic platelet reactivity, modifiable by dual antiplatelet therapy (DAPT), and the extrinsic inhibitory endothelial mediators, nitric oxide (NO) and prostacyclin (PGI2 ), that are powerfully potentiated by P2Y12 receptor blockade. This implies that for individual patients endothelial mediator production is an important determinant of DAPT effectiveness. Here, we have investigated this idea using platelets taken from healthy volunteers treated with anti-platelet drugs. METHODS: Three groups of male volunteers (n = 8) received either prasugrel (10 mg), aspirin (75 mg) or DAPT (prasugrel + aspirin) once daily for 7 days. Platelet reactivity in the presence of diethylammonium (Z)-1-(N,N-diethylamino)diazen-1-ium-1,2-diolate (DEA/NONOate) and PGI2 was studied before and following treatment. RESULTS: Ex vivo, PGI2 and/or DEA/NONOate had little inhibitory effect on TRAP-6-induced platelet reactivity in control conditions. However, in the presence of DAPT, combination of DEA/NONOate + PGI2 reduced platelet aggregation (74 ± 3% to 19 ± 6%, P < 0.05). In vitro studies showed even partial (25%) P2Y12 receptor blockade produced a significant (67 ± 2% to 39 ± 10%, P < 0.05) inhibition when DEA/NONOate + PGI2 was present. CONCLUSIONS: We have demonstrated that PGI2 and NO synergize with P2Y12 receptor antagonists to produce powerful platelet inhibition. Furthermore, even with submaximal P2Y12 blockade the presence of PGI2 and NO greatly enhances platelet inhibition. Our findings highlight the importance of endothelial mediator in vivo modulation of P2Y12 inhibition and introduces the concept of refining ex vivo platelet function testing by incorporating an assessment of endothelial function to predict thrombotic outcomes better and adjust therapy to prevent adverse outcomes in individual patients.


Asunto(s)
Aspirina/farmacología , Epoprostenol/farmacología , Óxido Nítrico/farmacología , Activación Plaquetaria/efectos de los fármacos , Clorhidrato de Prasugrel/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Adolescente , Adulto , Aspirina/administración & dosificación , Plaquetas/efectos de los fármacos , Sinergismo Farmacológico , Epoprostenol/administración & dosificación , Epoprostenol/metabolismo , Voluntarios Sanos , Humanos , Técnicas In Vitro , Masculino , Óxido Nítrico/administración & dosificación , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Agregación Plaquetaria/efectos de los fármacos , Clorhidrato de Prasugrel/administración & dosificación , Antagonistas del Receptor Purinérgico P2Y/administración & dosificación , Adulto Joven
3.
Proc Natl Acad Sci U S A ; 110(39): 15782-7, 2013 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-24003163

RESUMEN

Circulating platelets are constantly exposed to nitric oxide (NO) released from the vascular endothelium. This NO acts to reduce platelet reactivity, and in so doing blunts platelet aggregation and thrombus formation. For successful hemostasis, platelet activation and aggregation must occur at sites of vascular injury despite the constant presence of NO. As platelets aggregate, they release secondary mediators that drive further aggregation. Particularly significant among these secondary mediators is ADP, which, acting through platelet P2Y12 receptors, strongly amplifies aggregation. Platelet P2Y12 receptors are the targets of very widely used antithrombotic drugs such as clopidogrel, prasugrel, and ticagrelor. Here we show that blockade of platelet P2Y12 receptors dramatically enhances the antiplatelet potency of NO, causing a 1,000- to 100,000-fold increase in inhibitory activity against platelet aggregation and release reactions in response to activation of receptors for either thrombin or collagen. This powerful synergism is explained by blockade of a P2Y12 receptor-dependent, NO/cGMP-insensitive phosphatidylinositol 3-kinase pathway of platelet activation. These studies demonstrate that activation of the platelet ADP receptor, P2Y12, severely blunts the inhibitory effects of NO. The powerful antithrombotic effects of P2Y12 receptor blockers may, in part, be mediated by profound potentiation of the effects of endogenous NO.


Asunto(s)
Óxido Nítrico/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y12/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/enzimología , Proteína C-Reactiva/farmacología , Calcio/metabolismo , Moléculas de Adhesión Celular/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Epoprostenol/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Humanos , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Trombina/farmacología , Proteínas de Unión al GTP rap1/metabolismo
4.
Proc Natl Acad Sci U S A ; 109(43): 17597-602, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23045674

RESUMEN

Prostacyclin is an antithrombotic hormone produced by the endothelium, whose production is dependent on cyclooxygenase (COX) enzymes of which two isoforms exist. It is widely believed that COX-2 drives prostacyclin production and that this explains the cardiovascular toxicity associated with COX-2 inhibition, yet the evidence for this relies on indirect evidence from urinary metabolites. Here we have used a range of experimental approaches to explore which isoform drives the production of prostacyclin in vitro and in vivo. Our data show unequivocally that under physiological conditions it is COX-1 and not COX-2 that drives prostacyclin production in the cardiovascular system, and that urinary metabolites do not reflect prostacyclin production in the systemic circulation. With the idea that COX-2 in endothelium drives prostacyclin production in healthy individuals removed, we must seek new answers to why COX-2 inhibitors increase the risk of cardiovascular events to move forward with drug discovery and to enable more informed prescribing advice.


Asunto(s)
Sistema Cardiovascular/metabolismo , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Epoprostenol/biosíntesis , Animales , Western Blotting , Células Cultivadas , Ciclooxigenasa 1/genética , Ciclooxigenasa 2/genética , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Endotelio Vascular/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
FASEB J ; 27(10): 3938-46, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23792301

RESUMEN

Inhibition of cyclooxygenase (COX)-2 increases cardiovascular deaths. Identifying a biomarker of COX-2 is desirable but difficult, since COX-1 and COX-2 ordinarily catalyze formation of an identical product, prostaglandin H2. When acetylated by aspirin, however, COX-2 (but not COX-1) can form 15(R)-HETE, which is metabolized to aspirin-triggered lipoxin (ATL), 15-epi-lipoxin A4. Here we have used COX-1- and COX-2-knockout mice to establish whether plasma ATL could be used as a biomarker of vascular COX-2 in vivo. Vascular COX-2 was low but increased by LPS (10 mg/kg; i.p). Aspirin (10 mg/kg; i.v.) inhibited COX-1, measured as blood thromboxane and COX-2, measured as lung PGE2. Aspirin also increased the levels of ATL in the lungs of LPS-treated wild-type C57Bl6 mice (vehicle: 25.5±9.3 ng/ml; 100 mg/kg: 112.0±7.4 ng/ml; P<0.05). Despite this, ATL was unchanged in plasma after LPS and aspirin. This was true in wild-type as well as COX-1(-/-) and COX-2(-/-) mice. Thus, in mice in which COX-2 has been induced by LPS treatment, aspirin triggers detectable 15-epi-lipoxin A4 in lung tissue, but not in plasma. This important study is the first to demonstrate that while ATL can be measured in tissue, plasma ATL is not a biomarker of vascular COX-2 expression.


Asunto(s)
Aspirina/farmacología , Ciclooxigenasa 2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/toxicidad , Lipoxinas/metabolismo , Pulmón/enzimología , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Aspirina/administración & dosificación , Biomarcadores , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/genética , Relación Dosis-Respuesta a Droga , Lipoxinas/genética , Pulmón/efectos de los fármacos , Ratones
6.
Arterioscler Thromb Vasc Biol ; 31(2): 384-91, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21127291

RESUMEN

OBJECTIVE: The goal of this study was to examine the effect of chronic heterogeneous shear stress, applied using an orbital shaker, on endothelial cell morphology and the expression of cyclooxygenases 1 and 2. METHODS AND RESULTS: Porcine aortic endothelial cells were plated on fibronectin-coated Transwell plates. Cells were cultured for up to 7 days either under static conditions or on an orbital shaker that generated a wave of medium inducing shear stress over the cells. Cells were fixed and stained for the endothelial surface marker CD31 or cyclooxygenases 1 and 2. En face confocal microscopy and scanning ion conductance microscopy were used to show that endothelial cells were randomly oriented at the center of the well, aligned with shear stress nearer the periphery, and expressed cyclooxygenase-1 under all conditions. Lipopolysaccharide induced cyclooxygenase-2 and the production of 6-keto-prostaglandin F(1α) in all cells. CONCLUSIONS: Cyclooxygenase-1 is expressed in endothelial cells cultured under chronic shear stress of high or low directionality.


Asunto(s)
Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Estrés Mecánico , 6-Cetoprostaglandina F1 alfa/metabolismo , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Recuento de Células , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Isoenzimas/metabolismo , Lipopolisacáridos/farmacología , Modelos Animales , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Porcinos
7.
Biochem Biophys Res Commun ; 408(4): 511-5, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21513697

RESUMEN

Toll like receptor (TLR)4 is a pattern recognition receptor expressed in endothelial and other cells, responsible for the sensing of endotoxin and host derived ligands. Our group has shown previously that the absence of TLR4 is associated with reduced endothelial dependent vasodilator responses and left heart hypertrophy in animal models. However, the mechanism behind reduced endothelial cell function in TLR4(-/-) mice is not known. We have used en face confocal imaging of mesenteric arteries from mice deficient in the TLR4 receptor stained with dihydroethidium (DHE) to measure superoxide production. Using the isometric wire myograph, mesenteric artery vasodilator responses to acetylcholine and MnCl(2) (a superoxide dismutase mimetic) were measured. Mesenteric arteries from TLR4(-/-) mice had a reduced endothelial dependent relaxant response and increased superoxide levels when stimulated with acetylcholine. Increased levels of superoxide, as detected by DHE staining, were seen in vessels from TLR4(-/-) mice, which were reduced to control levels in the presence of MnCl(2). Our observations suggest that loss of TLR4 increases superoxide generation which reduces the biological activity of endothelial derived nitric oxide and thereby explains the endothelial dysfunction and associated cardiovascular phenotype in TLR4(-/-) mice. These data implicate a novel cardio-protective role for TLR4 in vascular homeostasis.


Asunto(s)
Vasos Sanguíneos/metabolismo , Endotelio Vascular/metabolismo , Superóxidos/metabolismo , Receptor Toll-Like 4/fisiología , Vasodilatación , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Acetilcolina/farmacología , Animales , Vasos Sanguíneos/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Factores Relajantes Endotelio-Dependientes/farmacología , Femenino , Ratones , Ratones Mutantes , Óxido Nítrico/farmacología , Receptor Toll-Like 4/genética , Vasoconstrictores/farmacología
8.
Vascul Pharmacol ; 71: 70-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25869509

RESUMEN

Regular consumption of chocolate and cocoa products has been linked to reduced cardiovascular mortality. This study compared the effects of high flavanol dark chocolate (HFDC; 1064mg flavanols/day for 6weeks) and low flavanol dark chocolate (LFDC; 88mg flavanols/day for 6weeks) on blood pressure, heart rate, vascular function and platelet aggregation in men with pre-hypertension or mild hypertension. Vascular function was assessed by pulse wave analysis using radial artery applanation tonometry in combination with inhaled salbutamol (0.4mg) to assess changes due to endothelium-dependent vasodilatation. HFDC did not significantly reduce blood pressure compared to baseline or LFDC. Heart rate was increased by LFDC compared to baseline, but not by HFDC. Vascular responses to salbutamol tended to be greater after HFDC. Platelet aggregation induced by collagen or the thromboxane analogue U46619 was unchanged after LFDC or HFDC, whereas both chocolates reduced responses to ADP and the thrombin receptor activator peptide, SFLLRNamide (TRAP6), relative to baseline. Pre-incubation of platelets with theobromine also attenuated platelet aggregation induced by ADP or TRAP6. We conclude that consumption of HFDC confers modest improvements in cardiovascular function. Platelet aggregation is modulated by a flavanol-independent mechanism that is likely due to theobromine.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Cacao , Endotelio Vascular/efectos de los fármacos , Flavonoides/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Presión Sanguínea/fisiología , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Endotelio Vascular/fisiología , Estudios de Seguimiento , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Persona de Mediana Edad , Agregación Plaquetaria/fisiología , Resultado del Tratamiento , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
9.
PLoS One ; 9(6): e98165, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24887395

RESUMEN

Cyxlo-oxygenase (COX)-2 inhibitors, including traditional nonsteroidal anti-inflammatory drugs (NSAIDs) are associated with increased cardiovascular side effects, including myocardial infarction. We and others have shown that COX-1 and not COX-2 drives vascular prostacyclin in the healthy cardiovascular system, re-opening the question of how COX-2 might regulate cardiovascular health. In diseased, atherosclerotic vessels, the relative contribution of COX-2 to prostacyclin formation is not clear. Here we have used apoE(-/-)/COX-2(-/-) mice to show that, whilst COX-2 profoundly limits atherosclerosis, this protection is independent of local prostacyclin release. These data further illustrate the need to look for new explanations, targets and pathways to define the COX/NSAID/cardiovascular risk axis. Gene expression profiles in tissues from apoE(-/-)/COX-2(-/-) mice showed increased lymphocyte pathways that were validated by showing increased T-lymphocytes in plaques and elevated plasma Th1-type cytokines. In addition, we identified a novel target gene, rgl1, whose expression was strongly reduced by COX-2 deletion across all examined tissues. This study is the first to demonstrate that COX-2 protects vessels against atherosclerotic lesions independently of local vascular prostacyclin and uses systems biology approaches to identify new mechanisms relevant to development of next generation NSAIDs.


Asunto(s)
Aterosclerosis/enzimología , Vasos Sanguíneos/metabolismo , Ciclooxigenasa 2/metabolismo , Epoprostenol/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Sustancias Protectoras/metabolismo , Linfocitos T/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Aterosclerosis/patología , Vasos Sanguíneos/patología , Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/genética , Femenino , Eliminación de Gen , Regulación Enzimológica de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Transducción de Señal , Transcriptoma/genética , Regulación hacia Arriba/genética
10.
PLoS One ; 8(7): e69524, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874970

RESUMEN

There are two schools of thought regarding the cyclooxygenase (COX) isoform active in the vasculature. Using urinary prostacyclin markers some groups have proposed that vascular COX-2 drives prostacyclin release. In contrast, we and others have found that COX-1, not COX-2, is responsible for vascular prostacyclin production. Our experiments have relied on immunoassays to detect the prostacyclin breakdown product, 6-keto-PGF1α and antibodies to detect COX-2 protein. Whilst these are standard approaches, used by many laboratories, antibody-based techniques are inherently indirect and have been criticized as limiting the conclusions that can be drawn. To address this question, we measured production of prostanoids, including 6-keto-PGF1α, by isolated vessels and in the circulation in vivo using liquid chromatography tandem mass spectrometry and found values essentially identical to those obtained by immunoassay. In addition, we determined expression from the Cox2 gene using a knockin reporter mouse in which luciferase activity reflects Cox2 gene expression. Using this we confirm the aorta to be essentially devoid of Cox2 driven expression. In contrast, thymus, renal medulla, and regions of the brain and gut expressed substantial levels of luciferase activity, which correlated well with COX-2-dependent prostanoid production. These data are consistent with the conclusion that COX-1 drives vascular prostacyclin release and puts the sparse expression of Cox2 in the vasculature in the context of the rest of the body. In doing so, we have identified the thymus, gut, brain and other tissues as target organs for consideration in developing a new understanding of how COX-2 protects the cardiovascular system.


Asunto(s)
Vasos Sanguíneos/metabolismo , Ciclooxigenasa 1/genética , Ciclooxigenasa 2/genética , Epoprostenol/metabolismo , Transcriptoma , 6-Cetoprostaglandina F1 alfa/metabolismo , Animales , Aorta/metabolismo , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados , Especificidad de Órganos/genética , Prostaglandinas/metabolismo , Espectrometría de Masas en Tándem
11.
PLoS One ; 7(2): e31228, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22359578

RESUMEN

OBJECTIVE: To measure the elongation and compliance of endothelial cells subjected to different patterns of shear stress in vitro, and to compare these parameters with the elongation and compliance of endothelial cells from different regions of the intact aorta. MATERIALS AND METHODS: Porcine aortic endothelial cells were cultured for 6 days under static conditions or on an orbital shaker. The shaker generated a wave of medium, inducing pulsatile shear stress with a preferred orientation at the edge of the well or steadier shear stress with changing orientation at its centre. The topography and compliance of these cells and cells from the inner and outer curvature of ex vivo porcine aortic arches were measured by scanning ion conductance microscopy (SICM). RESULTS: Cells cultured under oriented shear stress were more elongated and less compliant than cells grown under static conditions or under shear stress with no preferred orientation. Cells from the outer curvature of the aorta were more elongated and less compliant than cells from the inner curvature. CONCLUSION: The elongation and compliance of cultured endothelial cells vary according to the pattern of applied shear stress, and are inversely correlated. A similar inverse correlation occurs in the aortic arch, with variation between regions thought to experience different haemodynamic stresses.


Asunto(s)
Aorta/citología , Células Endoteliales/citología , Animales , Forma de la Célula , Células Cultivadas , Hemodinámica , Iones/metabolismo , Microscopía , Estrés Mecánico , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA