Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cardiovasc Drugs Ther ; 34(1): 15-23, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32062793

RESUMEN

PURPOSE: Drugs inhibiting the platelet P2Y12 receptor, such as clopidogrel and prasugrel, are potent antithrombotic agents and are widely used in cardiovascular disease. However, the adverse effects of these drugs have limited their clinical use. For example, clopidogrel resistance occurs in approximately one third of patients, while prasugrel increases the risk of major bleeding. Therefore, new generations of such drugs are of clinical interest. METHODS: In this study, the pharmacodynamics of a new P2Y12 antagonist, CN-218, was compared with that of clopidogrel and prasugrel in rats and mice. The differences between CN-218 and clopidogrel include deuteration of the 7-position methyl carboxylate and the introduction of cinnamate in the 2-position of thiophene. RESULTS: CN-218 had an antiaggregatory efficacy that was at least five times more potent than that of clopidogrel but not as potent as that of prasugrel. It had a significant impact on activated partial thromboplastin time (APTT), whereby the APTT of CN-218-treated rats was approximately 9 s longer than that of the vehicle- or clopidogrel-treated group, while it had no impact on prothrombin time (PT) in rats. CN-218 had a similar potent antithrombotic effect to that of prasugrel and clopidogrel and also reduced the risk of bleeding compared to prasugrel. CONCLUSION: CN-218 may be a promising antithrombotic agent, with potent antiplatelet and significant anticoagulant activity, as well as a lower risk of bleeding compared to clopidogrel and prasugrel.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Fibrinolíticos/farmacología , Piperidinas/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y12/efectos de los fármacos , Tiofenos/farmacología , Trombosis/prevención & control , Animales , Plaquetas/metabolismo , Carragenina , Clopidogrel/farmacología , AMP Cíclico/sangre , Modelos Animales de Enfermedad , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Masculino , Ratones , Piperidinas/toxicidad , Inhibidores de Agregación Plaquetaria/toxicidad , Clorhidrato de Prasugrel/farmacología , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Ratas Wistar , Receptores Purinérgicos P2Y12/sangre , Tiofenos/toxicidad , Trombosis/sangre , Trombosis/inducido químicamente
2.
Pharmacol Rep ; 70(1): 156-160, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29367102

RESUMEN

BACKGROUND: Though prasugrel is one of the important P2Y12 inhibitors currently in use for antiplatelet therapy, its potential effects on contractility and electrical activity of ventricular myocytes have not yet been investigated. Hence this study was designed to study the impact of prasugrel on contractile function and membrane potential of isolated ventricular myocytes. METHODS: Freshly isolated rat ventricular myocytes were used in this study. Myocyte contraction was measured during electrical stimulation of cardiomyocytes and the action potential (AP) recordings were obtained with current clamp mode of the patch-clamp amplifier. RESULTS: AP duration and fractional shortening of ventricular myocytes did not show any change with the administration of 1µM of prasugrel. However, remarkable depolarization of resting membrane potential followed by apparent fibrillation episodes was detected in the cardiomyocytes. Similar events were observed in the contractile activity of myocytes during field stimulation. Also, a higher concentration of prasugrel (10µM) elicited repeated fibrillations, which disappeared after washout or nitric oxide synthase (NOS) inhibition with L-NAME. In contrast, the same concentration of ticagrelor, another P2Y12 inhibitor did not induce fibrillation events though it decreased the contractility of ventricular myocytes significantly. The perfusion of ventricular myocytes with L-NAME did not alter the negative inotropic effect of ticagrelor. CONCLUSIONS: Prasugrel, a widely used antithrombotic agent, may induce depolarization in the membrane potential of myocytes as well as fibrillation via NO mediated pathway.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Fibrilación Atrial/inducido químicamente , Ventrículos Cardíacos/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Clorhidrato de Prasugrel/toxicidad , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Función Ventricular/efectos de los fármacos , Animales , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Miocitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Ratas Wistar , Factores de Tiempo
3.
Cell Physiol Biochem ; 44(4): 1337-1351, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29183030

RESUMEN

BACKGROUND/AIMS: Extracellular ATP performs multiple important functions via activation of P2 receptors on the cell surface. P2Y receptors play critical roles in ATP evoked response in human lung adenocarcinoma cells (A549 cells). Emodin is an anthraquinone derivative originally isolated from Chinese rhubarb, possesses anticancer properties. In this study we examined the inhibiting effects of emodin on proliferation, migration and epithelial-mesenchymal transition (EMT) by suppressing P2Y receptors-dependent Ca2+ increase and nuclear factor-κB (NF-KB) signaling in A549 cells. METHODS: A549 cells were pretreated with emodin before stimulation with ATP for the indicated time. Then, intracellular Ca2+ concentration ([Ca2+]i) was measured by Fluo-8/AM staining. Cell proliferation and cell cycle progression were tested by CCK8 assay and flow cytometry In addition, wound healing and western blot were performed to determine cell migration and related protein levels (Bcl-2, Bax, claudin-1, NF-κB). RESULTS: Emodin blunted ATP/UTP-induced increase of [Ca2+]i and cell proliferation concentration-dependently Meanwhile, it decreased ATP-induced cells accumulation in the S phase. Furthermore, emodin altered protein abundance of Bcl-2, Bax and claudin-1 and attenuated EMT caused by ATP. Such ATP-induced cellular reactions were also inhibited by a nonselective P2Y receptors antagonist, suramin, in a similar way to emodin. Besides, emodin could inhibit activation of NF-κB, thus suppressed ATP-induced proliferation, migration and EMT. CONCLUSION: Our results demonstrated that emodin inhibits ATP-induced proliferation, migration, EMT by suppressing P2Y receptors-mediated [Ca2+]i increase and NF-κB signaling in A549 cells.


Asunto(s)
Adenosina Trifosfato/farmacología , Proliferación Celular/efectos de los fármacos , Emodina/toxicidad , Receptores Purinérgicos P2Y/metabolismo , Transducción de Señal/efectos de los fármacos , Células A549 , Adenocarcinoma , Adenocarcinoma del Pulmón , Cadherinas/metabolismo , Calcio/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Claudina-1/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Neoplasias Pulmonares , Microscopía Fluorescente , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Receptores Purinérgicos P2Y/química , Receptores Purinérgicos P2Y/genética , Suramina/toxicidad , Proteína X Asociada a bcl-2/metabolismo
4.
Thromb Haemost ; 117(8): 1651-1659, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28569920

RESUMEN

The efficiency of current dual antiplatelet therapy might be further improved by its combination with a glycoprotein (GP) VI-targeting strategy without increasing bleeding. GPVI-Fc, a recombinant dimeric fusion protein binding to plaque collagen and concealing binding sites for platelet GPVI, acts as a lesion-focused antiplatelet drug, and does not increase bleeding in vivo. We investigated, whether GPVI-Fc added in vitro on top of acetylsalicylic acid (ASA), the P2Y12 antagonist ticagrelor, and the fibrinogen receptor antagonist abciximab alone or in combination would increase inhibition of platelet activation by atherosclerotic plaque. Under static conditions, GPVI-Fc inhibited plaque-induced platelet aggregation by 53 %, and increased platelet inhibition by ASA (51 %) and ticagrelor (64 %) to 66 % and 80 %, respectively. Under arterial flow, GPVI-Fc inhibited plaque-induced platelet aggregation by 57 %, and significantly increased platelet inhibition by ASA (28 %) and ticagrelor (47 %) to about 81 % each. The triple combination of GPVI-Fc, ASA and ticagrelor achieved almost complete inhibition of plaque-induced platelet aggregation (93 %). GPVI-Fc alone or in combination with ASA or ticagrelor did not increase closure time measured by the platelet function analyzer (PFA)-200. GPVI-Fc added on top of abciximab, a clinically used anti-fibrinogen receptor antibody which blocks platelet aggregation, strongly inhibited total (81 %) and stable (89 %) platelet adhesion. We conclude that GPVI-Fc added on top of single or dual antiplatelet therapy with ASA and/or a P2Y12 antagonist is likely to improve anti-atherothrombotic protection without increasing bleeding risk. In contrast, the strong inhibition of platelet adhesion by GPVI-Fc in combination with GPIIb/IIIa inhibitors could be harmful.


Asunto(s)
Adenosina/análogos & derivados , Anticuerpos Monoclonales/farmacología , Aspirina/farmacología , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Glicoproteínas/farmacología , Fragmentos Fab de Inmunoglobulinas/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Placa Aterosclerótica , Inhibidores de Agregación Plaquetaria/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Trombosis/prevención & control , Abciximab , Adenosina/farmacología , Adenosina/toxicidad , Anticuerpos Monoclonales/toxicidad , Aspirina/toxicidad , Plaquetas/metabolismo , Quimioterapia Combinada , Glicoproteínas/toxicidad , Hemorragia/inducido químicamente , Humanos , Fragmentos Fab de Inmunoglobulinas/toxicidad , Fragmentos Fc de Inmunoglobulinas/toxicidad , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/toxicidad , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Trombosis/sangre , Trombosis/patología , Ticagrelor , Factores de Tiempo
5.
Arterioscler Thromb Vasc Biol ; 35(3): 637-44, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25593131

RESUMEN

OBJECTIVE: Platelet hyperactivity is associated with vascular disease and contributes to the genesis of thrombotic disorders. ADP plays an important role in platelet activation and activates platelets through 2 G-protein-coupled receptors, the Gq-coupled P2Y1 receptor (P2Y1R), and the Gi-coupled P2Y12 receptor. Although the involvement of the P2Y1R in thrombogenesis is well established, there are no antagonists that are currently available for clinical use. APPROACH AND RESULTS: Our goal is to determine whether a novel antibody targeting the ligand-binding domain, ie, second extracellular loop (EL2) of the P2Y1R (EL2Ab) could inhibit platelet function and protect against thrombogenesis. Our results revealed that the EL2Ab does indeed inhibit ADP-induced platelet aggregation, in a dose-dependent manner. Furthermore, EL2Ab was found to inhibit integrin GPIIb-IIIa activation, dense and α granule secretion, and phosphatidylserine exposure. These inhibitory effects translated into protection against thrombus formation, as evident by a prolonged time for occlusion in a FeCl3-induced thrombosis model, but this was accompanied by a prolonged tail bleeding time. We also observed a dose-dependent displacement of the radiolabeled P2Y1R antagonist [(3)H]MRS2500 from its ligand-binding site by EL2Ab. CONCLUSIONS: Collectively, our findings demonstrate that EL2Ab binds to and exhibits P2Y1R-dependent function-blocking activity in the context of platelets. These results add further evidence for a role of the P2Y1R in thrombosis and validate the concept that targeting it is a relevant alternative or complement to current antiplatelet strategies.


Asunto(s)
Anticuerpos/farmacología , Plaquetas/efectos de los fármacos , Fibrinolíticos/farmacología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y1/efectos de los fármacos , Animales , Anticuerpos/metabolismo , Anticuerpos/toxicidad , Sitios de Unión , Unión Competitiva , Plaquetas/metabolismo , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Nucleótidos de Desoxiadenina/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Epítopos , Fibrinolíticos/metabolismo , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Hemostasis/efectos de los fármacos , Humanos , Ligandos , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilserinas/sangre , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/metabolismo , Inhibidores de Agregación Plaquetaria/toxicidad , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Antagonistas del Receptor Purinérgico P2Y/metabolismo , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Receptores Purinérgicos P2Y1/sangre , Receptores Purinérgicos P2Y1/deficiencia , Receptores Purinérgicos P2Y1/genética , Receptores Purinérgicos P2Y1/inmunología , Receptores Purinérgicos P2Y1/metabolismo , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Trombosis/sangre , Trombosis/prevención & control , Factores de Tiempo
6.
Thromb Res ; 134(3): 693-703, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25064036

RESUMEN

P2Y12 antagonism is a key therapeutic strategy in the management and prevention of arterial thrombosis. The objective of this study was to characterize the pharmacodynamic (PD) and pharmacokinetic (PK) properties of SAR216471, a novel P2Y12 receptor antagonist. SAR216471 blocks the binding of 2MeSADP to P2Y12 receptors in vitro (IC50=17 nM). This inhibition was shown to be reversible. It potently antagonized ADP-induced platelet aggregation in human and rat platelet-rich plasma (IC50=108 and 62 nM, respectively). It also inhibited platelet aggregation when blood was exposed to collagen or thromboxane A2. Its high selectivity was demonstrated against a large panel of receptors, enzymes, and ion channels. Despite its moderate bioavailability in rats, oral administration of SAR216471 resulted in a fast, potent, and sustained inhibition of platelet aggregation where the extent and duration of platelet inhibition were directly proportional to its circulating plasma levels. Pre-clinical study of SAR216471 in a rat shunt thrombosis model demonstrated a dose-dependent antithrombotic activity after oral administration (ED50=6.7 mg/kg). By comparison, ED50 values for clopidogrel, prasugrel and ticagrelor were 6.3, 0.35 and 2.6 mg/kg, respectively. Finally, the anti-hemostatic effect of SAR216471 and its competitors was investigated in a rat tail bleeding model, revealing a favorable safety profile of SAR216471. Together, these findings have established a reliable antiplatelet profile of SAR216471, and support its potential use in clinical practice as an alternative to currently available P2Y12 receptor antagonists.


Asunto(s)
Plaquetas/efectos de los fármacos , Fibrinolíticos/farmacología , Indoles/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2Y/farmacología , Piridazinas/farmacología , Receptores Purinérgicos P2Y12/efectos de los fármacos , Receptores Purinérgicos P2/efectos de los fármacos , Trombosis/prevención & control , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/metabolismo , Administración Oral , Animales , Unión Competitiva , Disponibilidad Biológica , Plaquetas/metabolismo , Células CHO , Cricetulus , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Fibrinolíticos/administración & dosificación , Fibrinolíticos/farmacocinética , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Humanos , Indoles/administración & dosificación , Indoles/farmacocinética , Indoles/toxicidad , Masculino , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacocinética , Inhibidores de Agregación Plaquetaria/toxicidad , Unión Proteica , Antagonistas del Receptor Purinérgico P2Y/administración & dosificación , Antagonistas del Receptor Purinérgico P2Y/farmacocinética , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Piridazinas/administración & dosificación , Piridazinas/farmacocinética , Piridazinas/toxicidad , Ratas Sprague-Dawley , Receptores Purinérgicos P2/sangre , Receptores Purinérgicos P2Y12/sangre , Transducción de Señal/efectos de los fármacos , Tionucleótidos/metabolismo , Trombosis/sangre , Transfección
7.
Respir Physiol Neurobiol ; 189(3): 498-505, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23994825

RESUMEN

Newborn mammals exhibit a biphasic hypoxic ventilatory response (HVR), but the relative contributions of carotid body-initiated CNS mechanisms versus central hypoxia on ventilatory depression during the late phase of the HVR are not well understood. Neonatal rats (P4-5 or P13-15) were treated with a nonselective P2 purinergic receptor antagonist (pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid, or PPADS; 125mgkg(-1), i.p.) to pharmacologically denervate the peripheral chemoreceptors. At P4-5, rats reared in normoxia showed a progressive decline in ventilation during a 10-min exposure to 12% O2 (21-28% decrease from baseline). No hypoxic ventilatory depression was observed in the older group of neonatal rats (i.e., P13-15), suggesting that the contribution of central hypoxia to hypoxic ventilatory depression diminishes with age. In contrast, rats reared in moderate hyperoxia (60% O2) from birth exhibited no hypoxic ventilatory depression at either age studied. Systemic PPADS had no effect on the ventilatory response to 7% CO2, suggesting that the drug did not cross the blood-brain barrier. These findings indicate that (1) CNS hypoxia depresses ventilation in young, neonatal rats independent of carotid body activation and (2) hyperoxia alters the development of CNS pathways that modulate the late phase of the hypoxic ventilatory response.


Asunto(s)
Sistema Nervioso Central/fisiopatología , Hiperoxia/fisiopatología , Insuficiencia Respiratoria/fisiopatología , Mecánica Respiratoria/fisiología , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Desnervación Autonómica/métodos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Inhibidores de Agregación Plaquetaria , Ventilación Pulmonar/efectos de los fármacos , Ventilación Pulmonar/fisiología , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/toxicidad , Ratas , Ratas Sprague-Dawley , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/patología , Factores de Tiempo
8.
Br J Pharmacol ; 169(1): 82-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23347039

RESUMEN

BACKGROUND AND PURPOSE: Prasugrel is a third-generation thienopyridine prodrug and ticagrelor is a non-competitive P2Y12 receptor antagonist. In their phase 3 studies, both agents reduced rates of ischemic events relative to treatment with clopidogrel. EXPERIMENTAL APPROACH: The pharmacodynamic profile of anti-platelet effects of prasugrel was compared with that of ticagrelor in rats. KEY RESULTS: The active metabolite of prasugrel was less potent than ticagrelor and its active metabolite on platelet aggregation in vitro. In contrast, prasugrel was a more potent antiplatelet agent than ticagrelor on ex vivo platelet aggregation: their ED50 values at peak for ADP 20 µmol·L(-1) were 1.9 and 8.0 mg·kg(-1) , respectively. Prasugrel's inhibition of platelet aggregation was maintained for up to 24 h after administration, but ticagrelor's duration of action was substantially shorter. Prasugrel and ticagrelor significantly inhibited thrombus formation with ED50 values of 1.8 and 7.7 mg·kg(-1) , respectively. Both agents also prolonged bleeding times (ED200 values of 3.0 and 13 mg·kg(-1) respectively) suggesting that at equivalent levels of inhibition of platelet aggregation, the agents would show comparable antithrombotic activity with similar bleeding risk. Platelet transfusion significantly increased blood platelet numbers similarly in prasugrel- and ticagrelor-treated rats. In the prasugrel-treated group, platelet transfusion caused significant shortening of bleeding time, while in the ticagrelor-treated group, platelet transfusion showed no influence on bleeding time under the experimental conditions employed. CONCLUSIONS AND IMPLICATIONS: Prasugrel and ticagrelor showed several differences in their pharmacological profiles and these disparities may reflect their differing reversibility and/or pharmacokinetic profiles.


Asunto(s)
Adenosina/análogos & derivados , Piperazinas/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Tiofenos/farmacología , Adenosina/administración & dosificación , Adenosina/farmacología , Adenosina/toxicidad , Animales , Pruebas de Coagulación Sanguínea , Relación Dosis-Respuesta a Droga , Hemorragia/inducido químicamente , Hemostasis/efectos de los fármacos , Masculino , Piperazinas/administración & dosificación , Piperazinas/toxicidad , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/toxicidad , Clorhidrato de Prasugrel , Profármacos , Antagonistas del Receptor Purinérgico P2Y/administración & dosificación , Antagonistas del Receptor Purinérgico P2Y/farmacología , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Ratas , Ratas Sprague-Dawley , Riesgo , Tiofenos/administración & dosificación , Tiofenos/toxicidad , Trombosis/prevención & control , Ticagrelor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...