Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Diabetes Investig ; 12(12): 2172-2182, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34043882

RESUMEN

AIMS/INTRODUCTION: Thrombin exerts various pathophysiological functions by activating protease-activated receptors (PARs). Recent data have shown that PARs influence the development of glomerular diseases including diabetic kidney disease (DKD) by regulating inflammation. Heparin cofactor II (HCII) specifically inactivates thrombin; thus, we hypothesized that low plasma HCII activity correlates with DKD development, as represented by albuminuria. MATERIALS AND METHODS: Plasma HCII activity and spot urine biomarkers, including albumin and liver-type fatty acid-binding protein (L-FABP), were determined as the urine albumin-to-creatinine ratio (uACR) and the urine L-FABP-to-creatinine ratio (uL-FABPCR) in 310 Japanese patients with diabetes mellitus (176 males and 134 females). The relationships between plasma HCII activities and those DKD urine biomarkers were statistically evaluated. In addition, the relationship between plasma HCII activities and annual uACR changes was statistically evaluated for 201/310 patients (115 males and 86 females). RESULTS: The mean plasma HCII activity of all participants was 93.8 ± 17.7%. Multivariate-regression analysis including confounding factors showed that plasma HCII activity independently contributed to the suppression of the uACR and log-transformed uACR values (P = 0.036 and P = 0.006, respectively) but not uL-FABPCR (P = 0.541). In addition, plasma HCII activity significantly and inversely correlated with annual uACR and log-transformed uACR increments after adjusting for confounding factors (P = 0.001 and P = 0.014, respectively). CONCLUSIONS: The plasma HCII activity was inversely and specifically associated with glomerular injury in patients with diabetes. The results suggest that HCII can serve as a novel predictive factor for early-stage DKD development, as represented by albuminuria.


Asunto(s)
Albuminuria/sangre , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/orina , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/orina , Cofactor II de Heparina/análisis , Adulto , Anciano , Albúminas/metabolismo , Albuminuria/orina , Biomarcadores/sangre , Biomarcadores/orina , Creatinina/orina , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/etiología , Proteínas de Unión a Ácidos Grasos/orina , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptores Proteinasa-Activados/sangre , Análisis de Regresión , Trombina/metabolismo
2.
Int J Exp Pathol ; 100(1): 41-48, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30811756

RESUMEN

Diabetes is associated with an increased risk of cardiovascular disease. This is partially attributed to an altered activation status of blood platelets in this disease. Previously, alterations have been shown in COX-1 and protease activated receptor (PAR)-3 receptor expression in platelets in two animal models of diabetes, there have not been studies which address expression of these proteins in mice with long-term streptozotocin (STZ)-induced diabetes. We have also addressed the effect of diabetes on platelet adhesion under flow conditions. With the use of flow cytometry, we have shown that certain markers of platelet basal activation, such as active form of αIIb ß3 and of CD40L were increased in STZ-induced diabetic mice. Platelets from STZ-induced diabetic mice were also more reactive when stimulated with PAR-4 activating peptide as revealed by higher expression of active form of αIIb ß3 , membrane-bound on vWillebrand Factor and binding of exogenous fluorescein isothyanate-labelled fibrinogen. Expression of COX-1 and production of thromboxane A2 in platelets of STZ-induced diabetic mice were higher than in control animals. We observed no effect of diabetes on ability of platelets to form stable adhesions with fibrinogen in flow conditions. We conclude that although certain similarities exist between patterns of activation of platelets in animal models of diabetes, the differences should also be taken into account.


Asunto(s)
Plaquetas/enzimología , Ciclooxigenasa 1/sangre , Diabetes Mellitus Experimental/sangre , Proteínas de la Membrana/sangre , Adhesividad Plaquetaria , Estreptozocina , Animales , Ligando de CD40/sangre , Vasos Coronarios/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/enzimología , Epoprostenol/metabolismo , Masculino , Ratones Endogámicos C57BL , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Receptores Proteinasa-Activados/sangre , Tromboxano A2/sangre , Factor de von Willebrand/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 38(2): 344-352, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29146750

RESUMEN

OBJECTIVE: TRPM7 (transient receptor potential cation channel, subfamily M, member 7) is a ubiquitously expressed bifunctional protein comprising a transient receptor potential channel segment linked to a cytosolic α-type serine/threonine protein kinase domain. TRPM7 forms a constitutively active Mg2+ and Ca2+ permeable channel, which regulates diverse cellular processes in both healthy and diseased conditions, but the physiological role of TRPM7 kinase remains largely unknown. APPROACH AND RESULTS: Here we show that point mutation in TRPM7 kinase domain deleting the kinase activity in mice (Trpm7R/R ) causes a marked signaling defect in platelets. Trpm7R/R platelets showed an impaired PIP2 (phosphatidylinositol-4,5-bisphosphate) metabolism and consequently reduced Ca2+ mobilization in response to stimulation of the major platelet receptors GPVI (glycoprotein VI), CLEC-2 (C-type lectin-like receptor), and PAR (protease-activated receptor). Altered phosphorylation of Syk (spleen tyrosine kinase) and phospholipase C γ2 and ß3 accounted for these global platelet activation defects. In addition, direct activation of STIM1 (stromal interaction molecule 1) with thapsigargin revealed a defective store-operated Ca2+ entry mechanism in the mutant platelets. These defects translated into an impaired platelet aggregate formation under flow and protection of the mice from arterial thrombosis and ischemic stroke in vivo. CONCLUSIONS: Our results identify TRPM7 kinase as a key modulator of phospholipase C signaling and store-operated Ca2+ entry in platelets. The protection of Trpm7R/R mice from acute ischemic disease without developing intracranial hemorrhage indicates that TRPM7 kinase might be a promising antithrombotic target.


Asunto(s)
Arteriopatías Oclusivas/sangre , Plaquetas/metabolismo , Señalización del Calcio , Calcio/sangre , Infarto de la Arteria Cerebral Media/sangre , Canales Catiónicos TRPM/sangre , Trombosis/sangre , Animales , Arteriopatías Oclusivas/genética , Arteriopatías Oclusivas/patología , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/patología , Lectinas Tipo C/sangre , Ratones Mutantes , Fosfatidilinositol 4,5-Difosfato/sangre , Fosfolipasa C beta/sangre , Fosfolipasa C gamma/sangre , Fosforilación , Glicoproteínas de Membrana Plaquetaria/metabolismo , Mutación Puntual , Receptores Proteinasa-Activados/sangre , Molécula de Interacción Estromal 1/sangre , Sinaptofisina/sangre , Canales Catiónicos TRPM/deficiencia , Canales Catiónicos TRPM/genética , Trombosis/genética , Trombosis/patología
4.
Curr Res Transl Med ; 64(3): 135-139, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27765273

RESUMEN

Platelet activation is a complex balance of positive and negative signaling pathways. Several protein kinase C (PKC) isoforms are expressed in human platelets. They are a major regulator of platelet granule secretion, activation and aggregation activity. One of those isoforms is the PKCδ isozyme, it has a central yet complex role in platelets such as opposite signaling functions depending on the nature of the agonist, it concentration and pathway. In fact, it has been shown that PKCδ has an overall negative influence on platelet function in response to collagen, while, following PAR stimulation, PKCδ has a positive effect on platelet function. Understanding the crucial role of PKCδ in platelet functions is recently emerging in the literature, therefore, further investigations should shed light into its specific role in hemostasis. In this review, we focus on the different roles of PKCδ in platelet activation, aggregation and thrombus formation.


Asunto(s)
Coagulación Sanguínea/fisiología , Plaquetas/enzimología , Activación Plaquetaria/fisiología , Proteína Quinasa C-delta/fisiología , Animales , Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Colágeno/farmacología , Gránulos Citoplasmáticos/metabolismo , Humanos , Isoenzimas/sangre , Isoenzimas/química , Isoenzimas/fisiología , Ratones , Fosforilación , Activación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/metabolismo , Conformación Proteica , Dominios Proteicos , Proteína Quinasa C-delta/sangre , Proteína Quinasa C-delta/química , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Seudópodos/ultraestructura , Receptores Proteinasa-Activados/sangre , Transducción de Señal , Trombina/farmacología
5.
Arterioscler Thromb Vasc Biol ; 34(12): 2563-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25278288

RESUMEN

OBJECTIVE: Current antiplatelet strategies to prevent myocardial infarction and stroke are limited by bleeding risk. A better understanding of the roles of distinct platelet-activating pathways is needed. We determined whether platelet activation by 2 key primary activators, thrombin and collagen, plays distinct, redundant, or interacting roles in tail bleeding and carotid thrombosis in mice. APPROACH AND RESULTS: Platelets from mice deficient for the thrombin receptor protease-activated receptor-4 (Par4) and the collagen receptor glycoprotein VI protein (GPVI) lack responses to thrombin and collagen, respectively. We examined tail bleeding and FeCl3-induced carotid artery occlusion in mice lacking Par4, GPVI, or both. We also examined a series of Par mutants with increasing impairment of thrombin signaling in platelets. Ablation of thrombin signaling alone by Par4 deficiency increased blood loss in the tail bleeding assay and impaired occlusive thrombus formation in the carotid occlusion assay. GPVI deficiency alone had no effect. Superimposing GPVI deficiency on Par4 deficiency markedly increased effect size in both assays. In contrast to complete ablation of thrombin signaling, 9- and 19-fold increases in EC50 for thrombin-induced platelet activation had only modest effects. CONCLUSIONS: The observation that loss of Par4 uncovered large effects of GPVI deficiency implies that Par4 and GPVI made independent, partially redundant contributions to occlusive thrombus formation in the carotid and to hemostatic clot formation in the tail under the experimental conditions examined. At face value, these results suggest that thrombin- and collagen-induced platelet activation can play partially redundant roles, despite important differences in how these agonists are made available to platelets.


Asunto(s)
Trombosis de las Arterias Carótidas/sangre , Colágeno/sangre , Hemorragia/sangre , Activación Plaquetaria/fisiología , Trombina/metabolismo , Animales , Plaquetas/metabolismo , Trombosis de las Arterias Carótidas/etiología , Hemorragia/etiología , Hemostasis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteínas de Membrana Plaquetaria/deficiencia , Glicoproteínas de Membrana Plaquetaria/genética , Receptores Proteinasa-Activados/sangre , Receptores Proteinasa-Activados/deficiencia , Receptores Proteinasa-Activados/genética , Receptores de Trombina/sangre , Receptores de Trombina/deficiencia , Receptores de Trombina/genética , Cola (estructura animal)
6.
Br J Anaesth ; 113(6): 970-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25209096

RESUMEN

BACKGROUND: Drugs that act on the platelet P2Y12 receptor are responsible for postoperative bleeding in cardiac surgery. However, protease-activated receptor (PAR) that reacts to thrombin stimulation might still be active in patients treated with P2Y12 inhibitors. Preoperative platelet function testing could possibly guide the timing of surgery. We investigated the association between P2Y12 receptor and PAR inhibition and bleeding after cardiac surgery. METHODS: A retrospective cohort study of 361 patients undergoing cardiac surgery and treated with P2Y12 anti-platelet agents was undertaken. All patients received a preoperative multiplate electrode aggregometry testing of platelet P2Y12 receptor activity (ADPtest) and PAR reactivity with thrombin receptor-activating peptide (TRAP) stimulation. ADPtest and TRAPtest data measured before surgery were analysed for association with postoperative bleeding (ml per 12 h) and severe postoperative bleeding. RESULTS: Both the ADPtest and the TRAPtest were significantly (P=0.001) associated with postoperative bleeding. A threshold of 22 U for the ADPtest yielded a negative predictive value (NPV) of 94% and a positive predictive value (PPV) of 20%, and a threshold of 75 U for the TRAPtest yielded an NPV of 95% and a PPV of 23%. In the subgroup of patients with ADPtest <22 U, TRAPtest ≥75 U was not associated with severe bleeding (NPV of 100% and PPV of 37%). CONCLUSIONS: In patients taking P2Y12 receptor inhibitors, residual platelet reactivity to thrombin stimulation limits the risk of severe postoperative bleeding.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Inhibidores de Agregación Plaquetaria/efectos adversos , Hemorragia Posoperatoria/inducido químicamente , Antagonistas del Receptor Purinérgico P2Y/efectos adversos , Receptores de Trombina/antagonistas & inhibidores , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Agregación Plaquetaria , Pruebas de Función Plaquetaria , Hemorragia Posoperatoria/sangre , Valor Predictivo de las Pruebas , Cuidados Preoperatorios/métodos , Pronóstico , Receptores Proteinasa-Activados/sangre , Receptores Purinérgicos P2Y12/sangre , Estudios Retrospectivos , Medición de Riesgo/métodos
7.
Thromb Res ; 127 Suppl 2: S34-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21193113

RESUMEN

Inflammation and coagulation are two main host-defence systems that interact with each other. Inflammation activates coagulation and coagulation modulates the inflammatory activity in many ways. The contributing molecular pathways are reviewed. Thrombin and activated protein C (APC) and its receptor EPCR constitute a major physiological regulatory system to control vascular wall permeability during sepsis. Pro-inflammatory cellular effects of coagulation proteases as well as the anti-inflammatory effects of APC/EPCR are mediated by signaling via protease activated receptors PAR on mononuclear cells, endothelial cells, platelets, fibroblast, and smooth muscle cells. The beneficial effects of APC in sepsis are mainly dependent on the PAR-mediated cell-protective properties rather than the anticoagulant protease function on coagulation cofactors FV/Va and FVIII/VIIIa. Animal experiments with signaling selective APC-variants show promise in improving the therapeutic efficacy and safety of APC in sepsis.


Asunto(s)
Coagulación Sanguínea/fisiología , Inflamación/sangre , Animales , Antitrombinas/sangre , Fibrina/metabolismo , Humanos , Inflamación/fisiopatología , Mediadores de Inflamación/sangre , Proteína C/fisiología , Proteína C/uso terapéutico , Receptores Proteinasa-Activados/sangre , Sepsis/sangre , Sepsis/tratamiento farmacológico , Sepsis/fisiopatología
8.
Proc Natl Acad Sci U S A ; 107(43): 18605-10, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20930120

RESUMEN

Toward understanding their redundancies and interactions in hemostasis and thrombosis, we examined the roles of thrombin receptors (protease-activated receptors, PARs) and the ADP receptor P2RY12 (purinergic receptor P2Y G protein-coupled 12) in human and mouse platelets ex vivo and in mouse models. Par3(-/-) and Par4(+/-) mouse platelets showed partially decreased responses to thrombin, resembling those in PAR1 antagonist-treated human platelets. P2ry12(+/-) mouse platelets showed partially decreased responses to ADP, resembling those in clopidogrel-treated human platelets. Par3(-/-) mice showed nearly complete protection against carotid artery thrombosis caused by low FeCl(3) injury. Par4(+/-) and P2ry12(+/-) mice showed partial protection. Increasing FeCl(3) injury abolished such protection; combining partial attenuation of thrombin and ADP signaling, as in Par3(-/-):P2ry12(+/-) mice, restored it. Par4(-/-) mice, which lack platelet thrombin responses, showed still better protection. Our data suggest that (i) the level of thrombin driving platelet activation and carotid thrombosis was low at low levels of arterial injury and increased along with the contribution of thrombin-independent pathways of platelet activation with increasing levels of injury; (ii) although P2ry12 acts downstream of PARs to amplify platelet responses to thrombin ex vivo, P2ry12 functioned in thrombin/PAR-independent pathways in our in vivo models; and (iii) P2ry12 signaling was more important than PAR signaling in hemostasis models; the converse was noted for arterial thrombosis models. These results make predictions being tested by ongoing human trials and suggest hypotheses for new antithrombotic strategies.


Asunto(s)
Hemostasis/fisiología , Receptores Proteinasa-Activados/sangre , Receptores Purinérgicos P2Y12/sangre , Trombosis/sangre , Proteínas Adaptadoras Transductoras de Señales , Adenosina Difosfato/sangre , Adenosina Difosfato/farmacología , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Moléculas de Adhesión Celular/sangre , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Proteínas de Ciclo Celular , Femenino , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Receptores Proteinasa-Activados/deficiencia , Receptores Proteinasa-Activados/genética , Receptores Purinérgicos P2Y12/deficiencia , Receptores Purinérgicos P2Y12/genética , Receptores de Trombina/sangre , Receptores de Trombina/deficiencia , Receptores de Trombina/genética , Transducción de Señal , Trombina/metabolismo , Trombina/farmacología , Trombosis/etiología
9.
Arthritis Rheum ; 62(3): 869-77, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20131237

RESUMEN

OBJECTIVE: To investigate the expression of protease-activated receptors (PARs), their potential regulation by anticardiolipin antibodies (aCL), and their association with the expression of other molecules relevant to thrombosis in monocytes obtained from 62 patients with primary antiphospholipid syndrome (APS). METHODS: Monocytes were isolated from peripheral blood mononuclear cells by magnetic depletion of nonmonocytes. Expression of tissue factor (TF) and PARs 1-4 genes was measured by quantitative real-time reverse transcription-polymerase chain reaction. Cell surface TF and PARs 1-4 expression was analyzed by flow cytometry. For in vitro studies, purified normal monocytes were incubated with purified APS patient IgG, normal human serum IgG, or lipopolysaccharide, in the presence or absence of specific monoclonal antibodies anti-PAR-1 (ATAP2) or anti-PAR-2 (SAM11) to test the effect of blocking the active site of PAR-1 or PAR-2. RESULTS: Analysis of both mRNA and protein for the 4 PARs revealed significantly increased expression of PAR-2 as compared with the control groups. PAR-1 was significantly overexpressed in APS patients with thrombosis and in the control patients with thrombosis but without APS. PAR-3 expression was not significantly altered. PAR-4 expression was absent in all groups analyzed. In addition, we demonstrated a correlation between the levels of PAR-2 and the titers of IgG aCL, as well as parallel behavior of TF and PAR-2 expression. In vitro, IgG from APS patients significantly increased monocyte expression of PAR-1 and PAR-2. Inhibition studies suggested that there was direct cross-talk between TF and PAR-2, such that inhibition of PAR-2 prevented the aCL-induced expression of TF. CONCLUSION: These results provide the first demonstration of increased expression of PARs in monocytes from patients with APS. Thus, PAR antagonists might have therapeutic potential as antithrombotic agents in APS.


Asunto(s)
Síndrome Antifosfolípido/sangre , Monocitos/química , Receptores Proteinasa-Activados/sangre , Adulto , Anticuerpos Anticardiolipina/fisiología , Anticuerpos Monoclonales/inmunología , Síndrome Antifosfolípido/complicaciones , Femenino , Citometría de Flujo , Humanos , Inmunoglobulina G/aislamiento & purificación , Masculino , Persona de Mediana Edad , ARN/análisis , Receptor Cross-Talk/fisiología , Receptores Proteinasa-Activados/genética , Receptores Proteinasa-Activados/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tromboplastina/análisis , Trombosis/etiología
10.
Blood ; 112(1): 90-9, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18310501

RESUMEN

A fundamental property of platelets is their ability to transmit cytoskeletal contractile forces to extracellular matrices. While the importance of the platelet contractile mechanism in regulating fibrin clot retraction is well established, its role in regulating the primary hemostatic response, independent of blood coagulation, remains ill defined. Real-time analysis of platelet adhesion and aggregation on a collagen substrate revealed a prominent contractile phase during thrombus development, associated with a 30% to 40% reduction in thrombus volume. Thrombus contraction developed independent of thrombin and fibrin and resulted in the tight packing of aggregated platelets. Inhibition of the platelet contractile mechanism, with the myosin IIA inhibitor blebbistatin or through Rho kinase antagonism, markedly inhibited thrombus contraction, preventing the tight packing of aggregated platelets and undermining thrombus stability in vitro. Using a new intravital hemostatic model, we demonstrate that the platelet contractile mechanism is critical for maintaining the integrity of the primary hemostatic plug, independent of thrombin and fibrin generation. These studies demonstrate an important role for the platelet contractile mechanism in regulating primary hemostasis and thrombus growth. Furthermore, they provide new insight into the underlying bleeding diathesis associated with platelet contractility defects.


Asunto(s)
Plaquetas/fisiología , Fibrina/fisiología , Hemostasis/fisiología , Trombosis/sangre , Trombosis/etiología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Retracción del Coagulo/fisiología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Motoras Moleculares/fisiología , Miosina Tipo IIA no Muscular/antagonistas & inhibidores , Miosina Tipo IIA no Muscular/fisiología , Adhesividad Plaquetaria , Inhibidores de Proteínas Quinasas/farmacología , Receptores Proteinasa-Activados/sangre , Receptores Proteinasa-Activados/deficiencia , Receptores Proteinasa-Activados/genética , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/sangre
11.
Thromb Haemost ; 99(2): 305-15, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18278179

RESUMEN

Thrombin is a multifunctional serine protease generated at the site of vascular injury that transforms fibrinogen into fibrin, activates blood platelets and elicits multiple effects on a variety of cell types including endothelial cells, vascular smooth muscle cells (VSMC), monocytes, T lymphocytes and fibroblasts. Cellular effects of thrombin are mediated by protease-activated receptors (PARs), members of the G protein-coupled receptors that carry their own ligand which remains cryptic until unmasked by proteolytic cleavage. Thrombin signalling in platelets contributes to haemostasis and thrombosis. In normal arteries PARs are mainly expressed in endothelial cells, while their expression in VSMC is limited. Endothelial PARs participate in the regulation of vascular tone, vascular permeability and endothelial secretory activity while in VSMC they mediate contraction, migration, proliferation, hypertrophy and production of extracellular matrix. PARs contribute to the pro-inflammatory phenotype observed in endothelial dysfunction and their up-regulation in VSMC seems to be a key element in the pathogenesis of atherosclerosis and restenosis. In the last years a myriad of studies have emphasized the critical role of PAR signalling in thrombin mediated effects in haemostasis, inflammation, cancer and embryonic development. Lately, PARs have become a therapeutic target to inhibit platelet aggregation and thrombosis. Early data from a clinical trial (TRA-PCI) to evaluate safety and efficacy of a potent new oral thrombin receptor antagonist (TRA) have promisingly indicated that overall TRA treatment reduces adverse event rates without an increase in bleeding risk. In this paper we review cellular responses triggered by thrombin and their implication in vascular pathophysiology.


Asunto(s)
Aterosclerosis/metabolismo , Receptores Proteinasa-Activados/metabolismo , Transducción de Señal , Trombina/metabolismo , Trombosis/metabolismo , Animales , Aterosclerosis/sangre , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/fisiopatología , Coagulación Sanguínea , Plaquetas/metabolismo , Constricción Patológica , Células Endoteliales/metabolismo , Hemorragia/sangre , Hemorragia/metabolismo , Humanos , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica , Inhibidores de Agregación Plaquetaria/farmacología , Inhibidores de Agregación Plaquetaria/uso terapéutico , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores Proteinasa-Activados/sangre , Recurrencia , Transducción de Señal/efectos de los fármacos , Trombosis/sangre , Trombosis/tratamiento farmacológico , Trombosis/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...