Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cardiovasc Res ; 117(10): 2275-2288, 2021 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-32931562

RESUMEN

AIMS: Arterial thrombosis as a result of plaque rupture or erosion is a key event in acute cardiovascular events. Sirtuin 5 (SIRT5) belongs to the lifespan-regulating sirtuin superfamily and has been implicated in acute ischaemic stroke and cardiac hypertrophy. This project aims at investigating the role of SIRT5 in arterial thrombus formation. METHODS AND RESULTS: Sirt5 transgenic (Sirt5Tg/0) and knock-out (Sirt5-/-) mice underwent photochemically induced carotid endothelial injury to trigger arterial thrombosis. Primary human aortic endothelial cells (HAECs) were treated with SIRT5 silencing-RNA (si-SIRT5) as well as peripheral blood mononuclear cells from acute coronary syndrome (ACS) patients and non-ACS controls (case-control study, total n = 171) were used to increase the translational relevance of our data. Compared to wild-type controls, Sirt5Tg/0 mice displayed accelerated arterial thrombus formation following endothelial-specific damage. Conversely, in Sirt5-/- mice, arterial thrombosis was blunted. Platelet function was unaltered, as assessed by ex vivo collagen-induced aggregometry. Similarly, activation of the coagulation cascade as assessed by vascular and plasma tissue factor (TF) and TF pathway inhibitor expression was unaltered. Increased thrombus embolization episodes and circulating D-dimer levels suggested augmented activation of the fibrinolytic system in Sirt5-/- mice. Accordingly, Sirt5-/- mice showed reduced plasma and vascular expression of the fibrinolysis inhibitor plasminogen activator inhibitor (PAI)-1. In HAECs, SIRT5-silencing inhibited PAI-1 gene and protein expression in response to TNF-α. This effect was mediated by increased AMPK activation and reduced phosphorylation of the MAP kinase ERK 1/2, but not JNK and p38 as shown both in vivo and in vitro. Lastly, both PAI-1 and SIRT5 gene expressions are increased in ACS patients compared to non-ACS controls after adjustment for cardiovascular risk factors, while PAI-1 expression increased across tertiles of SIRT5. CONCLUSION: SIRT5 promotes arterial thrombosis by modulating fibrinolysis through endothelial PAI-1 expression. Hence, SIRT5 may be an interesting therapeutic target in the context of atherothrombotic events.


Asunto(s)
Traumatismos de las Arterias Carótidas/enzimología , Trombosis de las Arterias Carótidas/enzimología , Células Endoteliales/enzimología , Fibrinólisis , Sirtuinas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/enzimología , Adulto , Anciano , Animales , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/genética , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/genética , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Fosforilación , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Sirtuinas/genética
2.
Arterioscler Thromb Vasc Biol ; 41(2): 683-697, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33267663

RESUMEN

OBJECTIVE: Using 3KO (triple NOX [NADPH oxidase] knockout) mice (ie, NOX1-/-/NOX2-/-/NOX4-/-), we aimed to clarify the role of this family of enzymes in the regulation of platelets in vitro and hemostasis in vivo. Approach and Results: 3KO mice displayed significantly reduced platelet superoxide radical generation, which was associated with impaired platelet aggregation, adhesion, and thrombus formation in response to the key agonists collagen and thrombin. A comparison with single-gene knockouts suggested that the phenotype of 3KO platelets is the combination of the effects of the genetic deletion of NOX1 and NOX2, while NOX4 does not show any significant function in platelet regulation. 3KO platelets displayed significantly higher levels of cGMP-a negative platelet regulator that activates PKG (protein kinase G). The inhibition of PKG substantially but only partially rescued the defective phenotype of 3KO platelets, which are responsive to both collagen and thrombin in the presence of the PKG inhibitors KT5823 or Rp-8-pCPT-cGMPs, but not in the presence of the NOS (NO synthase) inhibitor L-NG-monomethyl arginine. In vivo, triple NOX deficiency protected against ferric chloride-driven carotid artery thrombosis and experimental pulmonary embolism, while hemostasis tested in a tail-tip transection assay was not affected. Procoagulatory activity of platelets (ie, phosphatidylserine surface exposure) and the coagulation cascade in platelet-free plasma were normal. CONCLUSIONS: This study indicates that inhibiting NOXs has strong antithrombotic effects partially caused by increased intracellular cGMP but spares hemostasis. NOXs are, therefore, pharmacotherapeutic targets to develop new antithrombotic drugs without bleeding side effects.


Asunto(s)
Coagulación Sanguínea , Plaquetas/enzimología , Trombosis de las Arterias Carótidas/enzimología , NADPH Oxidasas/sangre , Activación Plaquetaria , Embolia Pulmonar/enzimología , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/genética , Trombosis de las Arterias Carótidas/prevención & control , GMP Cíclico/sangre , Proteínas Quinasas Dependientes de GMP Cíclico/sangre , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Fibrinolíticos/farmacología , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Activación Plaquetaria/efectos de los fármacos , Embolia Pulmonar/sangre , Embolia Pulmonar/genética , Embolia Pulmonar/prevención & control , Transducción de Señal , Superóxidos/sangre
3.
Arterioscler Thromb Vasc Biol ; 41(2): 668-682, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33297751

RESUMEN

OBJECTIVE: Current antiplatelet medications increase the risk of bleeding, which leads to a clear clinical need in developing novel mechanism-based antiplatelet drugs. TYMP (Thymidine phosphorylase), a cytoplasm protein that is highly expressed in platelets, facilitates multiple agonist-induced platelet activation, and enhances thrombosis. Tipiracil hydrochloride (TPI), a selective TYMP inhibitor, has been approved by the Food and Drug Administration for clinical use. We tested the hypothesis that TPI is a safe antithrombotic medication. Approach and Results: By coexpression of TYMP and Lyn, GST (glutathione S-transferase) tagged Lyn-SH3 domain or Lyn-SH2 domain, we showed the direct evidence that TYMP binds to Lyn through both SH3 and SH2 domains, and TPI diminished the binding. TYMP deficiency significantly inhibits thrombosis in vivo in both sexes. Pretreatment of platelets with TPI rapidly inhibited collagen- and ADP-induced platelet aggregation. Under either normal or hyperlipidemic conditions, treating wild-type mice with TPI via intraperitoneal injection, intravenous injection, or gavage feeding dramatically inhibited thrombosis without inducing significant bleeding. Even at high doses, TPI has a lower bleeding side effect compared with aspirin and clopidogrel. Intravenous delivery of TPI alone or combined with tissue plasminogen activator dramatically inhibited thrombosis. Dual administration of a very low dose of aspirin and TPI, which had no antithrombotic effects when used alone, significantly inhibited thrombosis without disturbing hemostasis. CONCLUSIONS: This study demonstrated that inhibition of TYMP, a cytoplasmic protein, attenuated multiple signaling pathways that mediate platelet activation, aggregation, and thrombosis. TPI can be used as a novel antithrombotic medication without the increase in risk of bleeding.


Asunto(s)
Plaquetas/efectos de los fármacos , Trombosis de las Arterias Carótidas/prevención & control , Inhibidores Enzimáticos/farmacología , Fibrinolíticos/farmacología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Pirrolidinas/farmacología , Timidina Fosforilasa/antagonistas & inhibidores , Timina/farmacología , Animales , Aspirina/farmacología , Plaquetas/enzimología , Células COS , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/enzimología , Trombosis de las Arterias Carótidas/genética , Chlorocebus aethiops , Modelos Animales de Enfermedad , Terapia Antiplaquetaria Doble , Inhibidores Enzimáticos/toxicidad , Femenino , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/toxicidad , Unión Proteica , Pirrolidinas/toxicidad , Transducción de Señal , Timidina Fosforilasa/genética , Timidina Fosforilasa/metabolismo , Timina/toxicidad , Dominios Homologos src , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
4.
Eur J Clin Invest ; 50(2): e13191, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31797367

RESUMEN

BACKGROUND: Endothelial cells regulate the formation of blood clots; thus, genes selectively expressed in these cells could primarily determine thrombus formation. Apold1 (apolipoprotein L domain containing 1) is a gene expressed by endothelial cells; whether Apold1 directly contributes to arterial thrombosis has not yet been investigated. Here, we assessed the effect of Apold1 deletion on arterial thrombus formation using an in vivo model of carotid thrombosis induced by photochemical injury. MATERIAL AND METHODS: Apold1 knockout (Apold1-/- ) mice and wild-type (WT) littermates underwent carotid thrombosis induced by photochemical injury, and time to occlusion was recorded. Tissue factor (TF) activity and activation of mitogen-activated protein kinases (MAPKs) and phosphatidyl-inositol-3 kinase (PI3K)/Akt pathways were analysed by colorimetric assay and Western blotting in both Apold1-/- and WT mice. Finally, platelet reactivity was assessed using light transmission aggregometry. RESULTS: After photochemical injury, Apold1-/- mice exhibited shorter time to occlusion as compared to WT mice. Moreover, TF activity was increased in carotid arteries of Apold1-/- when compared to WT mice. Underlying mechanistic markers such as TF mRNA and MAPKs activation were unaffected in Apold1-/- mice. In contrast, phosphorylation of Akt was reduced in Apold1-/- as compared to WT mice. Additionally, Apold1-/- mice displayed increased platelet reactivity to stimulation with collagen compared with WT animals. CONCLUSIONS: Deficiency of Apold1 results in a prothrombotic phenotype, accompanied by increased vascular TF activity, decreased PI3K/Akt activation and increased platelet reactivity. These findings suggest Apold1 as an interesting new therapeutic target in the context of arterial thrombosis.


Asunto(s)
Trombosis de las Arterias Carótidas/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Agregación Plaquetaria/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tromboplastina/metabolismo , Animales , Plaquetas/efectos de los fármacos , Colágeno Tipo I/farmacología , Células Endoteliales/metabolismo , Colorantes Fluorescentes , Proteínas Inmediatas-Precoces/genética , Coagulación con Láser , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Procesos Fotoquímicos , Agregación Plaquetaria/efectos de los fármacos , Pruebas de Función Plaquetaria , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Rosa Bengala , Transducción de Señal , Tromboplastina/genética
5.
Blood Adv ; 3(8): 1272-1284, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30995985

RESUMEN

Deficiency of the Nox2 (gp91phox) catalytic subunit of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase is a genetic cause of X-linked chronic granulomatous disease, a condition in which patients are prone to infection resulting from the loss of oxidant production by neutrophils. Some studies have suggested a role for superoxide derived from Nox2 NADPH oxidase in platelet activation and thrombosis, but data are conflicting. Using a rigorous and comprehensive approach, we tested the hypothesis that genetic deficiency of Nox2 attenuates platelet activation and arterial thrombosis. Our study was designed to test the genotype differences within male and female mice. Using chloromethyl-dichlorodihydrofluorescein diacetate, a fluorescent dye, as well as high-performance liquid chromatography analysis with dihydroethidium as a probe to detect intracellular reactive oxygen species (ROS), we observed no genotype differences in ROS levels in platelets. Similarly, there were no genotype-dependent differences in levels of mitochondrial ROS. In addition, we did not observe any genotype-associated differences in platelet activation, adhesion, secretion, or aggregation in male or female mice. Platelets from chronic granulomatous disease patients exhibited similar adhesion and aggregation responses as platelets from healthy subjects. Susceptibility to carotid artery thrombosis in a photochemical injury model was similar in wild-type and Nox2-deficient male or female mice. Our findings indicate that Nox2 NADPH oxidase is not an essential source of platelet ROS or a mediator of platelet activation or arterial thrombosis in large vessels, such as the carotid artery.


Asunto(s)
Plaquetas/enzimología , Trombosis de las Arterias Carótidas , NADPH Oxidasa 2 , Activación Plaquetaria , Especies Reactivas de Oxígeno/metabolismo , Animales , Trombosis de las Arterias Carótidas/enzimología , Trombosis de las Arterias Carótidas/genética , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , NADPH Oxidasa 2/genética , NADPH Oxidasa 2/metabolismo
6.
Thromb Res ; 155: 58-64, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28499154

RESUMEN

INTRODUCTION: Kunitz proteinase inhibitor (KPI) domain-containing forms of the amyloid ß-protein precursor (AßPP) inhibit cerebral thrombosis. KPI domain-lacking forms of AßPP are abundant in brain. Regions of AßPP other than the KPI domain may also be involved with regulating cerebral thrombosis. To determine the contribution of the KPI domain to the overall function of AßPP in regulating cerebral thrombosis we generated a reactive center mutant that was devoid of anti-thrombotic activity and studied its anti-thrombotic function in vitro and in vivo. METHODS: To determine the extent of KPI function of AßPP in regulating cerebral thrombosis we generated a recombinant reactive center KPIR13I mutant devoid of anti-thrombotic activity. The anti-proteolytic and anti-coagulant properties of wild-type and R13I mutant KPI were investigated in vitro. Cerebral thrombosis of wild-type, AßPP knock out and AßPP/KPIR13I mutant mice was evaluated in experimental models of carotid artery thrombosis and intracerebral hemorrhage. RESULTS: Recombinant mutant KPIR13I domain was ineffective in the inhibition of pro-thrombotic proteinases and did not inhibit the clotting of plasma in vitro. AßPP/KPIR13I mutant mice were similarly deficient as AßPP knock out mice in regulating cerebral thrombosis in experimental models of carotid artery thrombosis and intracerebral hemorrhage. CONCLUSIONS: We demonstrate that the anti-thrombotic function of AßPP primarily resides in the KPI activity of the protein.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Coagulación Sanguínea , Trombosis Intracraneal/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animales , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/genética , Trombosis de las Arterias Carótidas/metabolismo , Hemorragia Cerebral/sangre , Hemorragia Cerebral/genética , Hemorragia Cerebral/metabolismo , Técnicas de Sustitución del Gen , Humanos , Trombosis Intracraneal/sangre , Trombosis Intracraneal/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Dominios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
Eur Heart J ; 38(18): 1426-1435, 2017 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-26705390

RESUMEN

AIMS: Epidemiological studies strongly suggest a link between stress, depression, and cardiovascular diseases (CVDs); the mechanistic correlation, however, is poorly understood. A single-nucleotide polymorphism in the BDNF gene (BDNFVal66Met), associated with depression and anxiety, has been proposed as a genetic risk factor for CVD. Using a knock-in mouse carrying the BDNFVal66Met human polymorphism, which phenocopies psychiatric-related symptoms found in humans, we investigated the impact of this SNP on thrombosis. METHODS AND RESULTS: BDNFMet/Met mice displayed a depressive-like phenotype concomitantly with hypercoagulable state and platelet hyperreactivity. Proteomic analysis of aorta secretome from BDNFMet/Met and wild-type (WT) mice showed differential expression of proteins involved in the coagulation and inflammatory cascades. The BDNF Met allele predisposed to carotid artery thrombosis FeCl3-induced and to death after collagen/epinephrine injection. Interestingly, transfection with BDNFMet construct induced a prothrombotic/proinflammatory phenotype in WT cells. SIRT1 activation, using resveratrol and/or CAY10591, prevented thrombus formation and restored the physiological levels of coagulation and of platelet markers in BDNFMet/Met mice and/or cells transfected with the Met allele. Conversely, inhibition of SIRT1 by sirtinol and/or by specific siRNA induced the prothrombotic/proinflammatory phenotype in WT mice and cells. Finally, we found that BDNF Met homozygosity is associated with increased risk of acute myocardial infarction (AMI) in humans. CONCLUSION: Activation of platelets, alteration in coagulation pathways, and changes in vessel wall protein expression in BDNFMet/Met mice recapitulate well the features occurring in the anxiety/depression condition. Furthermore, our data suggest that the BDNFVal66Met polymorphism contribute to the individual propensity for arterial thrombosis related to AMI.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Trastorno Depresivo/genética , Polimorfismo de Nucleótido Simple/genética , Trombosis/genética , Animales , Trastornos de Ansiedad/genética , Aorta/fisiología , Coagulación Sanguínea/genética , Arterias Carótidas/fisiología , Trombosis de las Arterias Carótidas/genética , Modelos Animales de Enfermedad , Femenino , Heterocigoto , Homocigoto , Humanos , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Isquemia Miocárdica/genética , Proteínas del Tejido Nervioso/metabolismo , Activación Plaquetaria/genética , Inhibidores de Agregación Plaquetaria/farmacología , Receptores de Superficie Celular/metabolismo , Resveratrol , Transducción de Señal/fisiología , Sirtuina 1/antagonistas & inhibidores , Estilbenos/farmacología
8.
Thromb Haemost ; 115(5): 969-78, 2016 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-26790499

RESUMEN

Adaptor proteins play a critical role in the assembly of signalling complexes after engagement of platelet receptors by agonists such as collagen, ADP and thrombin. Recently, using proteomics, the Dok (downstream of tyrosine kinase) adapter proteins were identified in human and mouse platelets. In vitro studies suggest that Dok-1 binds to platelet integrin ß3, but the underlying effects of Dok-1 on αIIbß3 signalling, platelet activation and thrombosis remain to be elucidated. In the present study, using Dok-1-deficient (Dok-1-/-) mice, we determined the phenotypic role of Dok-1 in αIIbß3 signalling. We found that platelets from Dok-1-/- mice displayed normal aggregation, activation of αIIbß3 (assessed by binding of JON/A), P-selectin surface expression (assessed by anti-CD62P), and soluble fibrinogen binding. These findings indicate that Dok-1 does not affect "inside-out" platelet signalling. Compared with platelets from wild-type (WT) mice, platelets from Dok-1-/- mice exhibited increased clot retraction (p < 0.05 vs WT), increased PLCγ2 phosphorylation, and enhanced spreading on fibrinogen after thrombin stimulation (p < 0.01 vs WT), demonstrating that Dok-1 negatively regulates αIIbß3 "outside-in" signalling. Finally, we found that Dok-1-/- mice exhibited significantly shortened bleeding times and accelerated carotid artery thrombosis in response to photochemical injury (p < 0.05 vs WT mice). We conclude that Dok-1 modulates thrombosis and haemostasis by negatively regulating αIIbß3 outside-in signalling.


Asunto(s)
Proteínas de Unión al ADN/sangre , Fosfoproteínas/sangre , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteínas de Unión al ARN/sangre , Trombosis/prevención & control , Animales , Tiempo de Sangría , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/genética , Trombosis de las Arterias Carótidas/prevención & control , Retracción del Coagulo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Fibrinógeno/metabolismo , Hemostasis , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Selectina-P/sangre , Fosfolipasa C gamma/sangre , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Activación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Transducción de Señal , Trombosis/sangre , Trombosis/genética
9.
Blood ; 125(4): 710-9, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25339356

RESUMEN

The precise mechanism for reduced thrombosis in prekallikrein null mice (Klkb1(-/-)) is unknown. Klkb1(-/-) mice have delayed carotid artery occlusion times on the rose bengal and ferric chloride thrombosis models. Klkb1(-/-) plasmas have long-activated partial thromboplastin times and defective contact activation-induced thrombin generation that partially corrects upon prolonged incubation. However, in contact activation-induced pulmonary thromboembolism by collagen/epinephrine or long-chain polyphosphate, Klkb1(-/-) mice, unlike F12(-/-) mice, do not have survival advantage. Klkb1(-/-) mice have reduced plasma BK levels and renal B2R mRNA. They also have increased expression of the renal receptor Mas and plasma prostacyclin. Increased prostacyclin is associated with elevated aortic vasculoprotective transcription factors Sirt1 and KLF4. Treatment of Klkb1(-/-) mice with the Mas antagonist A-779, COX-2 inhibitor nimesulide, or Sirt1 inhibitor splitomicin lowers plasma prostacyclin and normalizes arterial thrombosis times. Treatment of normal mice with the Mas agonist AVE0991 reduces thrombosis. Klkb1(-/-) mice have reduced aortic tissue factor (TF) mRNA, antigen, and activity. In sum, Klkb1(-/-) mice have a novel mechanism for thrombosis protection in addition to reduced contact activation. This pathway arises when bradykinin delivery to vasculature is compromised and mediated by increased receptor Mas, prostacyclin, Sirt1, and KLF4, leading to reduced vascular TF.


Asunto(s)
Trombosis de las Arterias Carótidas , Epoprostenol , Factores de Transcripción de Tipo Kruppel , Precalicreína , Proteínas Proto-Oncogénicas , Receptores Acoplados a Proteínas G , Tromboplastina , Angiotensina II/análogos & derivados , Angiotensina II/farmacología , Animales , Trombosis de las Arterias Carótidas/inducido químicamente , Trombosis de las Arterias Carótidas/genética , Trombosis de las Arterias Carótidas/metabolismo , Trombosis de las Arterias Carótidas/patología , Epoprostenol/biosíntesis , Epoprostenol/genética , Imidazoles/farmacología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Noqueados , Naftalenos/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Tiempo de Tromboplastina Parcial , Fragmentos de Péptidos/farmacología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Pironas/farmacología , ARN Mensajero , Receptor de Bradiquinina B2/biosíntesis , Receptor de Bradiquinina B2/genética , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/biosíntesis , Sirtuina 1/genética , Sulfonamidas/farmacología , Sinaptotagminas/biosíntesis , Sinaptotagminas/genética , Tromboplastina/antagonistas & inhibidores , Tromboplastina/biosíntesis , Tromboplastina/genética
10.
Pol Arch Med Wewn ; 124(9): 474-82, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25072406

RESUMEN

Atherothrombosis is a preventable and multifaceted pathological disorder whose pathogenesis involves a large number of biological pathways such as lipid and hormonal metabolism, inflammation, and hemostasis. Although it has been known for a long time that atherosclerosis has a sizable hereditary component, research in the field of genetics of cardiovascular disease is still ongoing, with doubts often outweighing certainties. A large amount of evidence gathered so far allows to identify at least 5 potential important pathways that can be specifically targeted by genetic studies--lipoprotein metabolism, inflammation, the renin-angiotensin-aldosterone system, platelet function, blood coagulation, and fibrinolysis. Owing to a large number of published studies that have investigated the role of genetic polymorphisms in the pathogenesis of atherothrombosis and its complications, in this review, we focused on data emerging from meta­analyses. The available evidence suggests that some selected polymorphisms in low­density lipoprotein metabolism, C­reactive protein, and blood coagulation (especially factor V Leiden, prothrombin G20210A polymorphism, and plasminogen activator inhibitor type 1 4G/5G polymorphism) deserve particular attention. Of note, however, it seems implausible that one single polymorphism will add much to the current approach of risk assessment based on conventional risk factors. A paradigm shift would hence be needed in the current approach to the genetics of atherothrombosis, wherein the investigation of entire pathways rather than assessment of single mutations will likely provide more useful information for complex conditions that involve large numbers of genes and are subjected to environmental regulation of gene expression and cellular phenotype.


Asunto(s)
Trombosis de las Arterias Carótidas/genética , Embolia por Colesterol/genética , Inflamación/genética , Metabolismo de los Lípidos/genética , Predisposición Genética a la Enfermedad , Humanos , Polimorfismo Genético , Factores de Riesgo
11.
Br J Pharmacol ; 171(22): 5032-48, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24923668

RESUMEN

BACKGROUND AND PURPOSE: Combined hormone replacement therapy with oestrogens plus the synthetic progestin medroxyprogesterone acetate (MPA) is associated with an increased risk of thrombosis. However, the mechanisms of this pro-thrombotic effect are largely unknown. The purpose of this study was to: (i) compare the pro-thrombotic effect of MPA with another synthetic progestin, norethisterone acetate (NET-A), (ii) determine if MPA's pro-thrombotic effect can be antagonized by the progesterone and glucocorticoid receptor antagonist mifepristone and (iii) elucidate underlying mechanisms by comparing aortic gene expression after chronic MPA with that after NET-A treatment. EXPERIMENTAL APPROACH: Female apolipoprotein E-deficient mice were ovariectomized and treated with placebo, MPA, a combination of MPA + mifepristone or NET-A for 90 days on a Western-type diet. Arterial thrombosis was measured in vivo in a photothrombosis model. Aortic gene expression was analysed using microarrays; GeneOntology and KEGG pathway analyses were conducted. KEY RESULTS: MPA's pro-thrombotic effects were prevented by mifepristone, while NET-A did not affect arterial thrombosis. Aortic gene expression analysis showed, for the first time, that gestagens induce similar effects on a set of genes potentially promoting thrombosis. However, in NET-A-treated mice other genes with potentially anti-thrombotic effects were also affected, which might counterbalance the effects of the pro-thrombotic genes. CONCLUSIONS AND IMPLICATIONS: The pro-thrombotic effects of synthetic progestins appear to be compound-specific, rather than representing a class effect of gestagens. Furthermore, the different thrombotic responses elicited by MPA and NET-A might be attributed to a more balanced, 'homeostatic' gene expression induced in NET-A- as compared with MPA-treated mice.


Asunto(s)
Aorta/efectos de los fármacos , Trombosis de las Arterias Carótidas/genética , Anticonceptivos Femeninos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Acetato de Medroxiprogesterona/farmacología , Noretindrona/análogos & derivados , Animales , Aorta/metabolismo , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Células Cultivadas , Vasos Coronarios/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones Noqueados , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Noretindrona/farmacología , Acetato de Noretindrona , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovariectomía , Reacción en Cadena de la Polimerasa
12.
Blood ; 123(24): 3828-31, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24771858

RESUMEN

Multiple myeloma confers a high risk for vascular thrombosis, a risk that is increased by treatment with immunomodulatory agents. Strikingly, inclusion of the proteasome inhibitor bortezomib reduces thrombotic risk, yet the molecular basis for this observation remains unknown. Here, we show that bortezomib prolongs thrombosis times in the carotid artery photochemical injury assay in normal mice. Cell-based studies show that bortezomib increases expression of the transcription factor Kruppel-like factor 2 (KLF2) in multiple cell types. Global postnatal overexpression of KLF2 (GL-K2-TG) increased time to thrombosis, and global postnatal deletion of KLF2 (GL-K2-KO) conferred an antiparallel effect. Finally, studies in GL-K2-KO mice showed that the thromboprotective effect of bortezomib is KLF2 dependent. These findings identify a transcriptional basis for the antithrombotic effects of bortezomib.


Asunto(s)
Ácidos Borónicos/farmacología , Trombosis de las Arterias Carótidas/prevención & control , Citoprotección/genética , Factores de Transcripción de Tipo Kruppel/fisiología , Pirazinas/farmacología , Animales , Bortezomib , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/genética , Trombosis de las Arterias Carótidas/patología , Células Cultivadas , Oclusión Coronaria/genética , Oclusión Coronaria/patología , Oclusión Coronaria/prevención & control , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tiempo de Coagulación de la Sangre Total
13.
Blood ; 121(19): 3962-9, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23515926

RESUMEN

Factor XI (fXI) is a homodimeric zymogen that is converted to a protease with 1 (1/2-fXIa) or 2 (fXIa) active subunits by factor XIIa (fXIIa) or thrombin. It has been proposed that the dimeric structure is required for normal fXI activation. Consistent with this premise, fXI monomers do not reconstitute fXI-deficient mice in a fXIIa-dependent thrombosis model. FXI activation by fXIIa or thrombin is a slow reaction that can be accelerated by polyanions. Phosphate polymers released from platelets (poly-P) can enhance fXI activation by thrombin and promote fXI autoactivation. Poly-P increased initial rates of fXI activation 30- and 3000-fold for fXIIa and thrombin, respectively. FXI monomers were activated more slowly than dimers by fXIIa in the presence of poly-P. However, this defect was not observed when thrombin was the activating protease, nor during fXI autoactivation. The data suggest that fXIIa and thrombin activate fXI by different mechanisms. FXIIa may activate fXI through a trans-activation mechanism in which the protease binds to 1 subunit of the dimer, while activating the other subunit. For activation by thrombin, or during autoactivation, the data support a cis-activation mechanism in which the activating protease binds to and activates the same fXI subunit.


Asunto(s)
Factor XI/química , Factor XI/metabolismo , Factor XIa/metabolismo , Animales , Trombosis de las Arterias Carótidas/genética , Trombosis de las Arterias Carótidas/metabolismo , Factor XI/genética , Deficiencia del Factor XI/genética , Deficiencia del Factor XI/metabolismo , Factor XIIa/química , Factor XIIa/metabolismo , Factor XIa/química , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína
14.
Thromb Haemost ; 108(3): 516-26, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22836883

RESUMEN

Identifying coagulation abnormalities in patients with combined bleeding and thrombosis history is clinically challenging. Our goal was to probe the complexity of dysregulated coagulation in humans by characterizing pathophysiologic mechanisms in a patient with both bleeding and thrombosis. The patient is a 56-year-old female with a history of haematomas, poor wound healing, and thrombosis (retinal artery occlusion and transient cerebral ischaemia). She had a normal activated partial thromboplastin time, prolonged thrombin and reptilase times, and decreased functional and antigenic fibrinogen levels, and was initially diagnosed with hypodysfibrinogenaemia. This diagnosis was supported by DNA analysis revealing a novel FGB mutation (c.656A>G) predicting a Q189R mutation in the mature chain that was present in the heterozygote state. However, turbidity analysis showed that purified fibrinogen polymerisation and degradation were indistinguishable from normal, and Bß chain subpopulations appeared normal by two-dimensional difference in-gel electrophoresis, indicating the mutated chain was not secreted. Interestingly, plasma thrombin generation testing revealed the patient's thrombin generation was higher than normal and could be attributed to elevated levels of factor VIII (FVIII, 163-225%). Accordingly, in an arterial injury model, hypofibrinogenaemic mice (Fgn(+/-)) infused with factor VIII demonstrated significantly shorter vessel occlusion times than saline-infused Fgn(+/-) mice. Together, these data associate the complex bleeding and thrombotic presentation with combined hypofibrinogenaemia plus plasma hypercoagulability. These findings suggest previous cases in which fibrinogen abnormalities have been associated with thrombosis may also be complicated by co-existing plasma hypercoagulability and illustrate the importance of "global" coagulation testing in patients with compound presentations.


Asunto(s)
Afibrinogenemia/genética , Factor VIII/análisis , Fibrinógeno/genética , Trastornos Hemorrágicos/genética , Mutación Missense , Mutación Puntual , Trombina/biosíntesis , Trombofilia/genética , Afibrinogenemia/sangre , Afibrinogenemia/complicaciones , Sustitución de Aminoácidos , Animales , Biopolímeros , Pruebas de Coagulación Sanguínea , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/genética , Modelos Animales de Enfermedad , Electroforesis en Gel Bidimensional , Factor VIII/toxicidad , Femenino , Fibrinógeno/química , Fibrinólisis , Eliminación de Gen , Trastornos Hemorrágicos/sangre , Trastornos Hemorrágicos/etiología , Heterocigoto , Humanos , Ataque Isquémico Transitorio/etiología , Ratones , Ratones Mutantes , Persona de Mediana Edad , Oclusión de la Arteria Retiniana/etiología , Trombofilia/sangre , Trombofilia/etiología
15.
J Immunol ; 188(2): 885-91, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22156595

RESUMEN

Bleeding disorders and thrombotic complications constitute a major cause of death and disability worldwide. Although it is known that the complement and coagulation systems interact, no studies have investigated the specific role or mechanisms of lectin-mediated coagulation in vivo. FeCl(3) treatment resulted in intra-arterial occlusive thrombogenesis within 10 min in wild-type (WT) and C2/factor B-null mice. In contrast, mannose-binding lectin (MBL)-null and MBL-associated serine protease (MASP)-1/-3 knockout (KO) mice had significantly decreased FeCl(3)-induced thrombogenesis. Reconstitution with recombinant human (rh) MBL restored FeCl(3)-induced thrombogenesis in MBL-null mice to levels comparable to WT mice, suggesting a significant role of the MBL/MASP complex for in vivo coagulation. Additionally, whole blood aggregation demonstrated increased MBL/MASP complex-dependent platelet aggregation. In vitro, MBL/MASP complexes were captured on mannan-coated plates, and cleavage of a chromogenic thrombin substrate (S2238) was measured. We observed no significant differences in S2238 cleavage between WT, C2/factor B-null, MBL-A(-/-), or MBL-C(-/-) sera; however, MBL-null or MASP-1/-3 KO mouse sera demonstrated significantly decreased S2238 cleavage. rhMBL alone failed to cleave S2238, but cleavage was restored when rMASP-1 was added to either MASP-1/-3 KO sera or rhMBL. Taken together, these findings indicate that MBL/MASP complexes, and specifically MASP-1, play a key role in thrombus formation in vitro and in vivo.


Asunto(s)
Coagulación Sanguínea , Trombosis de las Arterias Carótidas/enzimología , Lectina de Unión a Manosa de la Vía del Complemento , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/fisiología , Animales , Coagulación Sanguínea/inmunología , Trombosis de las Arterias Carótidas/inducido químicamente , Trombosis de las Arterias Carótidas/genética , Cloruros/toxicidad , Lectina de Unión a Manosa de la Vía del Complemento/genética , Modelos Animales de Enfermedad , Compuestos Férricos/toxicidad , Humanos , Inmunidad Innata/genética , Lectinas de Unión a Manosa/deficiencia , Lectinas de Unión a Manosa/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/efectos adversos , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/deficiencia , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Ratones , Trombina/fisiología
17.
Blood ; 117(14): 3929-37, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21297000

RESUMEN

Prolylcarboxypeptidase (PRCP) activates prekallikrein to plasma kallikrein, leading to bradykinin liberation, and degrades angiotensin II. We now identify PRCP as a regulator of blood vessel homeostasis. ß-Galactosidase staining in PRCP(gt/gt) mice reveals expression in kidney and vasculature. Invasive telemetric monitorings show that PRCP(gt/gt) mice have significantly elevated blood pressure. PRCP(gt/gt) mice demonstrate shorter carotid artery occlusion times in 2 models, and their plasmas have increased thrombin generation times. Pharmacologic inhibition of PRCP with Z-Pro-Prolinal or plasma kallikrein with soybean trypsin inhibitor, Pro-Phe-Arg-chloromethylketone or PKSI 527 also shortens carotid artery occlusion times. Aortic and renal tissues have uncoupled eNOS and increased reactive oxygen species (ROS) in PRCP(gt/gt) mice as detected by dihydroethidium or Amplex Red fluorescence or lucigenin luminescence. The importance of ROS is evidenced by the fact that treatment of PRCP(gt/gt) mice with antioxidants (mitoTEMPO, apocynin, Tempol) abrogates the hypertensive, prothrombotic phenotype. Mechanistically, our studies reveal that PRCP(gt/gt) aortas express reduced levels of Kruppel-like factors 2 and 4, thrombomodulin, and eNOS mRNA, suggesting endothelial cell dysfunction. Further, PRCP siRNA treatment of endothelial cells shows increased ROS and uncoupled eNOS and decreased protein C activation because of thrombomodulin inactivation. Collectively, our studies identify PRCP as a novel regulator of vascular ROS and homeostasis.


Asunto(s)
Carboxipeptidasas/genética , Trombosis de las Arterias Carótidas/genética , Hipertensión/genética , Interferencia de ARN/fisiología , Enfermedades Vasculares/genética , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiopatología , Carboxipeptidasas/antagonistas & inhibidores , Carboxipeptidasas/fisiología , Trombosis de las Arterias Carótidas/complicaciones , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Hipertensión/complicaciones , Hipertensión/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Interferente Pequeño/farmacología , Tiempo de Trombina , Factores de Tiempo , Enfermedades Vasculares/complicaciones , Enfermedades Vasculares/fisiopatología
18.
Eur J Vasc Endovasc Surg ; 38(1): 20-5, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19356953

RESUMEN

OBJECTIVES: Thrombomodulatory factors have been implicated in plaque instability. The aim was to examine the relationship between thrombomodulatory gene expression, timing of clinical events and plaque histology. DESIGN OF STUDY: Plaques were obtained from 40 consecutive patients undergoing carotid endarterectomy and divided into three groups (group 1, early symptomatic, within 1 month; group 2, late symptomatic, 1-6 months and group 3, asymptomatic). Total RNA was isolated to determine the expression of tissue plasminogen activator (t-PA), urokinase plasminogen activator (u-PA), plasminogen activator inhibitor-1 (PAI-1), tissue factor (TF), tissue factor pathway inhibitor (TFPI), thrombomodulin (TM), CD68 and vascular endothelial-cadherin (VE-Cadherin). RESULTS: Expression of t-PA, PAI-1, TF, TFPI, TM, CD68 and VE-cadherin were significantly increased in the early symptomatic group (p=0.019, 0.028, 0.018, 0.025, 0.038, 0.016 and 0.027 respectively), but the level of gene expression in the late symptomatic group was indistinguishable from the asymptomatic group. The incidence of plaque rupture and intraplaque haemorrhage was significantly increased in the early symptomatic groups (58% versus 18%/18% group 2/3, and 55% versus 6%/9% respectively, p<0.05 for both). CONCLUSIONS: Expression of thrombomodulatory genes is increased in unstable plaques, though levels after 1 month are comparable to asymptomatic plaques. This transient rise may influence plaque instability, and rapid resolution mirrors the clinical reduction in risk of further thrombo-embolic events.


Asunto(s)
Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Factores de Coagulación Sanguínea/genética , Cadherinas/genética , Arterias Carótidas/metabolismo , Trombosis de las Arterias Carótidas/genética , ADN/genética , Regulación de la Expresión Génica , Anciano , Anciano de 80 o más Años , Antígenos CD/biosíntesis , Antígenos de Diferenciación Mielomonocítica/biosíntesis , Factores de Coagulación Sanguínea/biosíntesis , Cadherinas/biosíntesis , Arterias Carótidas/cirugía , Trombosis de las Arterias Carótidas/metabolismo , Trombosis de las Arterias Carótidas/cirugía , Endarterectomía Carotidea , Endotelio Vascular/metabolismo , Inhibidores del Factor Xa , Femenino , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Humanos , Lipoproteínas/biosíntesis , Lipoproteínas/genética , Macrófagos , Masculino , Persona de Mediana Edad , Fenotipo , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Inhibidor 1 de Activador Plasminogénico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombomodulina/biosíntesis , Trombomodulina/genética , Tromboplastina/biosíntesis , Tromboplastina/genética , Activador de Tejido Plasminógeno/biosíntesis , Activador de Tejido Plasminógeno/genética
19.
Arterioscler Thromb Vasc Biol ; 28(12): 2231-8, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18974383

RESUMEN

BACKGROUND: In patients with coronary artery disease and reduced ejection fraction, amiodarone reduces mortality by decreasing sudden death. Because the latter may be triggered by coronary artery thrombosis as much as ventricular arrhythmias, amiodarone might interfere with tissue factor (TF) expression and thrombus formation. METHODS AND RESULTS: Clinically relevant plasma concentrations of amiodarone reduced TF activity and impaired carotid artery thrombus formation in a mouse photochemical injury model in vivo. PTT, aPTT, and tail bleeding time were not affected; platelet number was slightly decreased. In human endothelial and vascular smooth muscle cells, amiodarone inhibited tumor necrosis factor (TNF)-alpha and thrombin-induced TF expression as well as surface activity. Amiodarone lacking iodine and the main metabolite of amiodarone, N-monodesethylamiodarone, inhibited TF expression. Amiodarone did not affect mitogen-activated protein kinase activation, TF mRNA expression, and TF protein degradation. Metabolic labeling confirmed that amiodarone inhibited TF protein translation. CONCLUSIONS: Amiodarone impairs thrombus formation in vivo; in line with this, it inhibits TF protein expression and surface activity in human vascular cells. These pleiotropic actions occur within the range of amiodarone concentrations measured in patients, and thus may account at least in part for its beneficial effects in patients with coronary artery disease.


Asunto(s)
Amiodarona/farmacología , Trombosis de las Arterias Carótidas/metabolismo , Trombosis de las Arterias Carótidas/prevención & control , Tromboplastina/biosíntesis , Amiodarona/análogos & derivados , Animales , Antiarrítmicos/farmacología , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Traumatismos de las Arterias Carótidas/etiología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Trombosis de las Arterias Carótidas/genética , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tromboplastina/genética
20.
J Thromb Haemost ; 6(4): 684-91, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18339097

RESUMEN

BACKGROUND: Platelet glycoprotein (GP) Ib-IX-V supports platelet adhesion on damaged vascular walls by binding to von Willebrand factor (VWF). For several decades it has been recognized that the alpha-subunit of GP (GPIbalpha) also binds thrombin but the physiological relevance, if any, of this interaction was unknown. Previous studies have shown that a sulfated tyrosine 276 (Tyr276) is essential for thrombin binding to GPIbalpha. OBJECTIVES: This study investigated the in vivo relevance of GPIbalpha residue Tyr276 in hemostasis and thrombosis. METHODS: Transgenic mouse colonies expressing the normal human GPIbalpha subunit or a mutant human GPIbalpha containing a Phe substitution for Tyr276 (hTg(Y276F)) were generated. Both colonies were bred to mice devoid of murine GPIbalpha. RESULTS: Surface-expressed GPIbalpha levels and platelet counts were similar in both colonies. hTg(Y276F) platelets were significantly impaired in binding alpha-thrombin but displayed normal binding to type I fibrillar collagen and human VWF in the presence of ristocetin. In vivo thrombus formation as a result of chemical damage (FeCl(3)) demonstrated that hTg(Y276F) mice have a delayed time to occlusion followed by unstable blood flow indicative of embolization. In models of laser-induced injury, thrombi developing in hTg(Y276F) animals were also less stable. CONCLUSIONS: The results demonstrate that GPIbalpha residue Tyr276 is physiologically important, supporting stable thrombus formation in vivo.


Asunto(s)
Coagulación Sanguínea/fisiología , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Tirosina/fisiología , Sustitución de Aminoácidos , Animales , Tiempo de Sangría , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/inducido químicamente , Trombosis de las Arterias Carótidas/etiología , Trombosis de las Arterias Carótidas/genética , Cloruros , Colágeno Tipo I/metabolismo , Compuestos Férricos/toxicidad , Humanos , Rayos Láser/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Agregación Plaquetaria , Recuento de Plaquetas , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/fisiología , Mutación Puntual , Ristocetina/farmacología , Trombina/metabolismo , Factor de von Willebrand/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA