Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros











Intervalo de año de publicación
1.
Braz. J. Pharm. Sci. (Online) ; 58: e201089, 2022. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1420429

RESUMEN

Abstract Protease-activated receptors (PARs) are metabotropic G-protein-coupled receptors that are activated via proteolytic cleavage of a specific sequence of amino acids in their N-terminal region. PAR2 has been implicated in mediating allergic airway inflammation. This study aims to study the effect of PAR2 antagonist ENMD1068in lung inflammation and airway remodeling in experimental asthma. Allergic lung inflammation was induced in sensitized BALB/c mice through intranasal instillations of ovalbumin (OVA), and mice were pretreated with ENMD1068 1 hour before each OVA challenge. Bronchoalveolar lavage fluid (BALF) was collected, and the lungs were removed at different time intervals after OVA challenge to analyze inflammation, airway remodeling and airway hyperresponsiveness. Ovalbumin promoted leukocyte infiltration into BALF in a PAR2-dependent manner. ENMD1068 impaired eosinophil peroxidase (EPO) and myeloperoxidase (MPO) activity in the lung parenchyma into BALF and reduced the loss of dynamic pulmonary compliance, lung resistance in response to methacholine, mucus production, collagen deposition and chemokine (C-C motif) ligand 5 expression compared to those in OVA-challenged mice. We propose that proteases released after an allergen challenge may be crucial to the development of allergic asthma in mice, and PAR2 blockade may be useful as a new pharmacological approach for the treatment of airway allergic diseases.


Asunto(s)
Animales , Femenino , Ratones , Neumonía/patología , Receptor PAR-2/antagonistas & inhibidores , Receptores Proteinasa-Activados/antagonistas & inhibidores , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos
2.
Int J Mol Sci ; 22(7)2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33916310

RESUMEN

Epidemiological evidence shows that smoking causes a thrombophilic milieu that may play a role in the pathophysiology of chronic obstructive pulmonary disease (COPD) as well as pulmonary thromboembolism. The increased nicotine level induces a prothrombotic status and abnormal blood coagulation in smokers. Since several anticoagulants increase bleeding risk, alternative therapies need to be identified to protect against thrombosis without affecting hemostasis. Astragalin is a flavonoid present in persimmon leaves and green tea seeds and exhibits diverse activities of antioxidant and anti-inflammation. The current study investigated that astragalin attenuated smoking-induced pulmonary thrombosis and alveolar inflammation. In addition, it was explored that molecular links between thrombosis and inflammation entailed protease-activated receptor (PAR) activation and oxidative stress-responsive mitogen-activated protein kinase (MAPK)-signaling. BALB/c mice were orally administrated with 10-20 mg/kg astragalin and exposed to cigarette smoke for 8 weeks. For the in vitro study, 10 U/mL thrombin was added to alveolar epithelial A549 cells in the presence of 1-20 µM astragalin. The cigarette smoking-induced the expression of PAR-1 and PAR-2 in lung tissues, which was attenuated by the administration of ≥10 mg/kg astragalin. The oral supplementation of ≥10 mg/kg astragalin to cigarette smoke-challenged mice attenuated the protein induction of urokinase plasminogen activator, plasminogen activator inhibitor-1and tissue factor, and instead enhanced the induction of tissue plasminogen activator in lung tissues. The astragalin treatment alleviated cigarette smoke-induced lung emphysema and pulmonary thrombosis. Astragalin caused lymphocytosis and neutrophilia in bronchoalveolar lavage fluid due to cigarette smoke but curtailed infiltration of neutrophils and macrophages in airways. Furthermore, this compound retarded thrombin-induced activation of PAR proteins and expression of inflammatory mediators in alveolar cells. Treating astragalin interrupted PAR proteins-activated reactive oxygen species production and MAPK signaling leading to alveolar inflammation. Accordingly, astragalin may interrupt the smoking-induced oxidative stress-MAPK signaling-inflammation axis via disconnection between alveolar PAR activation and pulmonary thromboembolism.


Asunto(s)
Quempferoles/uso terapéutico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Embolia Pulmonar/prevención & control , Enfisema Pulmonar/prevención & control , Receptores Proteinasa-Activados/antagonistas & inhibidores , Animales , Fumar Cigarrillos/efectos adversos , Evaluación Preclínica de Medicamentos , Quempferoles/farmacología , Masculino , Ratones Endogámicos BALB C , Estrés Oxidativo , Embolia Pulmonar/etiología
3.
Eur J Pharmacol ; 869: 172875, 2020 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-31877279

RESUMEN

There is growing evidence for the contribution of the activated coagulation factor X (FXa) in the development of chronic inflammatory lung diseases. Therefore, we aimed to investigate effects of exogenous FXa on mitochondrial and metabolic function as well as the induction of inflammatory molecules in type II alveolar epithelial cells. Effects of FXa on epithelial cells were investigated in A549 cell line. Activation of extracellular signal-regulated kinase (ERK) and induction of inflammatory molecules were examined by immunoblot and gene expression analysis. Mitochondrial function was assessed by the measurement of oxygen consumption during maximal oxidative phosphorylation and quantitative determination of cardiolipin oxidation. Apoptosis was tested using a caspase 3 antibody. Metabolic activity and lactate dehydrogenase assay were applied for the detection of cellular viability. FXa activated ERK1/2 and induced an increase in the expression of pro-inflammatory cytokines, which was prevented by an inhibitor of FXa, edoxaban, or an inhibitor of protease-activated receptor 1, vorapaxar. Exposure to FXa caused mitochondrial alteration with restricted capacity for ATP generation, which was effectively prevented by edoxaban, vorapaxar and GB83 (inhibitor of protease-activated receptor 2). Of note, exposure to FXa did not initiate apoptosis in epithelial cells. FXa-dependent pro-inflammatory state and impairment of mitochondria did not reach the level of significance in lung epithelial cells. However, these effects might limit regenerative potency of lung epithelial cells, particular under clinical circumstances where lung injury causes exposure to clotting factors.


Asunto(s)
Células Epiteliales/metabolismo , Factor Xa/metabolismo , Inflamación/metabolismo , Mitocondrias/metabolismo , Receptores Proteinasa-Activados/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Dipéptidos/farmacología , Células Epiteliales/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Inhibidores del Factor Xa/farmacología , Humanos , Isoxazoles/farmacología , Mitocondrias/efectos de los fármacos , Piridinas/farmacología , Receptores Proteinasa-Activados/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Tiazoles/farmacología
4.
J Leukoc Biol ; 105(4): 729-740, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30690783

RESUMEN

Tissue-type plasminogen activator (tPA) activates fibrinolysis and also suppresses innate immune system responses to LPS in bone marrow-derived macrophages (BMDMs) and in vivo in mice. The objective of this study was to assess the activity of tPA as a regulator of macrophage physiology in the presence of plasmin. Enzymatically active and enzymatically inactive (EI) tPA appeared to comprehensively block the response to LPS in BMDMs, including expression of proinflammatory cytokines such as TNF-α and IL-1ß and anti-inflammatory cytokines such as IL-10 and IL-1 receptor antagonist. The activity of EI-tPA as an LPS response modifier was conserved in the presence of plasminogen. By contrast, in BMDMs treated with tPA and plasminogen or preactivated plasmin, in the presence or absence of LPS, increased proinflammatory cytokine expression was observed and tPA failed to reverse the response. Plasmin independently activated NF-κB, ERK1/2, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase in BMDMs, which is characteristic of proinflammatory stimuli. Plasmin-induced cytokine expression was blocked by ε-aminocaproic acid, aprotinin, and inhibitors of the known plasmin substrate, Protease-activated receptor-1 (PAR-1), but not by N-methyl-d-aspartate receptor inhibitor, which blocks the effects of tPA on macrophages. Cytokine expression by BMDMs treated with the PAR-1 agonist, TFLLR, was not inhibited by EI-tPA, possibly explaining why EI-tPA does not inhibit macrophage responses to plasmin and providing evidence for specificity in the ability of tPA to oppose proinflammatory stimuli. Regulation of innate immunity by the fibrinolysis system may reflect the nature of the stimulus and a balance between the potentially opposing activities of tPA and plasmin.


Asunto(s)
Fibrinolisina/metabolismo , Inflamación/patología , Lipopolisacáridos/metabolismo , Receptores Proteinasa-Activados/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Péptidos/farmacología , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/antagonistas & inhibidores , Transducción de Señal
5.
J Immunol Res ; 2017: 5193572, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28503577

RESUMEN

Proteinase-activated receptors 1 (PAR1) and 2 (PAR2) are the most highly expressed members of the PAR family in the periodontium. These receptors regulate periodontal inflammatory and repair processes through their activation by endogenous and bacterial enzymes. PAR1 is expressed by the periodontal cells such as human gingival fibroblasts, gingival epithelial cells, periodontal ligament cells, osteoblasts, and monocytic cells and can be activated by thrombin, matrix metalloproteinase 1 (MMP-1), MMP-13, fibrin, and gingipains from Porphyromonas gingivalis. PAR2 is expressed by neutrophils, osteoblasts, oral epithelial cells, and human gingival fibroblasts, and its possible activators in the periodontium are gingipains, neutrophil proteinase 3, and mast cell tryptase. The mechanisms through which PARs can respond to periodontal enzymes and result in appropriate immune responses have until recently been poorly understood. This review discusses recent findings that are beginning to identify a cardinal role for PAR1 and PAR2 on periodontal tissue metabolism.


Asunto(s)
Periodontitis/metabolismo , Periodontitis/fisiopatología , Periodoncio/metabolismo , Receptor PAR-1/metabolismo , Receptores Proteinasa-Activados/metabolismo , Adhesinas Bacterianas/metabolismo , Animales , Células Cultivadas , Cisteína Endopeptidasas/metabolismo , Células Epiteliales , Fibroblastos , Regulación de la Expresión Génica , Cisteína-Endopeptidasas Gingipaínas , Encía/citología , Encía/metabolismo , Humanos , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Ratones , Periodontitis/genética , Porphyromonas gingivalis , Receptor PAR-1/agonistas , Receptor PAR-1/antagonistas & inhibidores , Receptor PAR-1/genética , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores Proteinasa-Activados/genética
6.
J Thromb Haemost ; 15(4): 597-607, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28079978

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease with a 5-year mortality rate of > 50% and unknown etiology. Treatment options remain limited and, currently, only two drugs are available, i.e. nintedanib and pirfenidone. However, both of these antifibrotic agents only slow down the progression of the disease, and do not remarkably prolong the survival of IPF patients. Hence, the discovery of new therapeutic targets for IPF is crucial. Studies exploring the mechanisms that are involved in IPF have identified several possible targets for therapeutic interventions. Among these, blood coagulation factor receptors, i.e. protease-activated receptors (PARs), are key candidates, as these receptors mediate the cellular effects of coagulation factors and play central roles in influencing inflammatory and fibrotic responses. In this review, we will focus on the controversial role of the coagulation cascade in the pathogenesis of IPF. In the light of novel data, we will attempt to reconciliate the apparently conflicting data and discuss the possibility of pharmacologic targeting of PARs for the treatment of fibroproliferative diseases.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Fibrosis Pulmonar Idiopática/metabolismo , Receptores Proteinasa-Activados/metabolismo , Animales , Anticoagulantes , Bleomicina/química , Factores de Coagulación Sanguínea/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis/patología , Humanos , Fibrosis Pulmonar Idiopática/mortalidad , Inflamación , Lactonas/uso terapéutico , Ratones , Inhibidores de Agregación Plaquetaria/uso terapéutico , Piridinas/uso terapéutico , Receptor PAR-1/antagonistas & inhibidores , Receptor PAR-2/antagonistas & inhibidores , Receptores Proteinasa-Activados/antagonistas & inhibidores
7.
Annu Rev Pharmacol Toxicol ; 57: 349-373, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27618736

RESUMEN

Protease-activated receptors (PARs) are a unique class of G protein-coupled receptors (GPCRs) that transduce cellular responses to extracellular proteases. PARs have important functions in the vasculature, inflammation, and cancer and are important drug targets. A unique feature of PARs is their irreversible proteolytic mechanism of activation that results in the generation of a tethered ligand that cannot diffuse away. Despite the fact that GPCRs have proved to be the most successful class of druggable targets, the development of agents that target PARs specifically has been challenging. As a consequence, researchers have taken a remarkable diversity of approaches to develop pharmacological entities that modulate PAR function. Here, we present an overview of the diversity of therapeutic agents that have been developed against PARs. We further discuss PAR biased signaling and the influence of receptor compartmentalization, posttranslational modifications, and dimerization, which are important considerations for drug development.


Asunto(s)
Descubrimiento de Drogas/métodos , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores Proteinasa-Activados/metabolismo , Animales , Descubrimiento de Drogas/tendencias , Humanos , Oligopéptidos/metabolismo , Oligopéptidos/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
8.
Toxicology ; 365: 9-16, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27475285

RESUMEN

Emerging evidence supports a protective effect of platelets in experimental cholestatic liver injury and cholangiofibrosis. Coagulation-mediated platelet activation has been shown to inhibit experimental chronic cholestatic liver necrosis and biliary fibrosis. This occurs through thrombin-mediated activation of protease activated receptor-4 (PAR-4) in mice. However, it is not known whether other pathways of platelet activation, such as adenosine diphosphate (ADP)-mediated receptor P2Y12 activation is also protective. We tested the hypothesis that inhibition of P2Y12-mediated platelet activation exacerbates hepatic injury and cholangiofibrosis, and examined the impact of P2Y12 inhibition in both the presence and absence of PAR-4. Treatment of wild-type mice with the P2Y12 receptor antagonist clopidogrel increased biliary hyperplasia and cholangiofibrosis in wild-type mice exposed to the xenobiotic alpha-naphthylisothiocyanate (ANIT) for 4 weeks compared to vehicle-treated mice exposed to ANIT. Interestingly, this effect of clopidogrel occurred without a corresponding increase in hepatocellular necrosis. Whereas biliary hyperplasia and cholangiofibrosis were increased in PAR-4(-/-) mice, clopidogrel treatment failed to further increase these pathologies in PAR-4(-/-) mice. The results indicate that inhibition of receptor P2Y12-mediated platelet activation exacerbates bile duct fibrosis in ANIT-exposed mice, independent of hepatocellular necrosis. Moreover, the lack of an added effect of clopidogrel administration on the exaggerated pathology in ANIT-exposed PAR-4(-/-) mice reinforces the prevailing importance of coagulation-mediated platelet activation in limiting this unique liver pathology.


Asunto(s)
Colestasis/patología , Cirrosis Hepática/patología , Activación Plaquetaria/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Proteinasa-Activados/antagonistas & inhibidores , Ticlopidina/análogos & derivados , 1-Naftilisotiocianato , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Colestasis/inducido químicamente , Clopidogrel , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Proteinasa-Activados/metabolismo , Serotonina/sangre , Ticlopidina/farmacología , Xenobióticos
9.
Thromb Res ; 136(6): 1216-23, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26412559

RESUMEN

INTRODUCTION: Dual anti-platelet therapy (DAPT) with aspirin and a P2Y12 antagonist is standard of care to reduce risk of thrombosis, but does not directly target thrombin-dependent platelet activation. Therefore, PAR-1 antagonist addition to DAPT (i.e., triple anti-platelet therapy; TAPT) may improve the efficacy of treatment, though at the expense of an increase in bleeding risk. Using an in vitro transfusion model, we evaluated if platelet function loss associated with TAPT can be remedied by the addition of drug-naïve platelets. METHODS: To mimic TAPT, platelet-rich plasma (PRP) prepared from consented DAPT patients (DPRP) was incubated with a vorapaxar at therapeutic plasma levels (TPRP). To simulate platelet transfusions, TPRP was mixed with increasing proportions of drug-naïve PRP (NPRP). Platelet function recovery was assessed by light transmission aggregometry (LTA), aggregate morphology, and P-selectin expression. RESULTS: LTA results demonstrated that 20% NPRP was required to restore the ADP aggregation response in TPRP to the response observed in DPRP and 40% NPRP recovered aggregation to >65%. Higher NPRP fractions (60%) were required to restore the platelet reactivity using TRAP-6 (SFLLRN) or arachidonic acid (AA). PAR-4 aggregation was unaffected by platelet antagonists. A decrease in single, free platelets and incorporation of mepacrine-labeled naïve platelets into aggregates occurred with increasing NPRP portions. Upon agonist activation, the surface density and percent of P-selectin positive platelets increased linearly upon addition of NPRP. CONCLUSION: This in vitro model demonstrated that administration of drug-naïve platelets can be a useful strategy for reversing overall platelet inhibition observed with TAPT.


Asunto(s)
Transfusión Sanguínea/métodos , Inhibidores de Agregación Plaquetaria/química , Agregación Plaquetaria/efectos de los fármacos , Pruebas de Función Plaquetaria , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Ácido Araquidónico/metabolismo , Aspirina/uso terapéutico , Plaquetas/efectos de los fármacos , Clopidogrel , Citometría de Flujo , Hemorragia , Humanos , Lactonas/uso terapéutico , Selectina-P/metabolismo , Fragmentos de Péptidos/metabolismo , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/uso terapéutico , Transfusión de Plaquetas , Antagonistas del Receptor Purinérgico P2Y/química , Piridinas/uso terapéutico , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores Purinérgicos P2Y12/metabolismo , Ticlopidina/análogos & derivados , Ticlopidina/uso terapéutico
10.
Arterioscler Thromb Vasc Biol ; 35(3): 500-12, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25633316

RESUMEN

Platelet G-protein-coupled receptors influence platelet function by mediating the response to various agonists, including ADP, thromboxane A2, and thrombin. Blockade of the ADP receptor, P2Y12, in combination with cyclooxygenase-1 inhibition by aspirin has been among the most widely used pharmacological strategies to reduce cardiovascular event occurrence in high-risk patients. The latter dual pathway blockade strategy is one of the greatest advances in the field of cardiovascular medicine. In addition to P2Y12, the platelet thrombin receptor, protease activated receptor-1, has also been recently targeted for inhibition. Blockade of protease activated receptor-1 has been associated with reduced thrombotic event occurrence when added to a strategy using P2Y12 and cyclooxygenase-1 inhibition. At this time, the relative contributions of these G-protein-coupled receptor signaling pathways to in vivo thrombosis remain incompletely defined. The observation of treatment failure in ≈10% of high-risk patients treated with aspirin and potent P2Y12 inhibitors provides the rationale for targeting novel pathways mediating platelet function. Targeting intracellular signaling downstream from G-protein-coupled receptor receptors with phosphotidylionisitol 3-kinase and Gq inhibitors are among the novel strategies under investigation to prevent arterial ischemic event occurrence. Greater understanding of the mechanisms of G-protein-coupled receptor-mediated signaling may allow the tailoring of antiplatelet therapy.


Asunto(s)
Plaquetas/efectos de los fármacos , Diseño de Fármacos , Terapia Molecular Dirigida , Inhibidores de Agregación Plaquetaria/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Trombosis/tratamiento farmacológico , Animales , Plaquetas/metabolismo , Humanos , Inhibidores de Agregación Plaquetaria/química , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores Proteinasa-Activados/metabolismo , Receptores Purinérgicos P2Y12/efectos de los fármacos , Receptores Purinérgicos P2Y12/metabolismo , Trombosis/sangre , Trombosis/metabolismo
11.
Nat Rev Cardiol ; 12(1): 30-47, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25286881

RESUMEN

For more than 10 years, dual antiplatelet therapy with aspirin and clopidogrel has remained the cornerstone of treatment for patients with acute coronary syndrome (ACS). The novel oral P2Y purinoceptor 12 (P2Y12)-receptor inhibitors prasugrel and ticagrelor were approved by the FDA for clinical use in 2009 and 2011, respectively. These agents have a faster-acting, more-potent, and more-predictable antiplatelet effect than clopidogrel, which translates into improved clinical outcomes in patients with ACS, albeit at the expense of an increased risk of bleeding. However, some patients continue to experience adverse ischaemic events despite treatment with aspirin and a P2Y12-receptor antagonist, because platelets can remain activated via pathways not inhibited by these agents, such as the protease-activated receptor (PAR)-1 platelet activation pathway stimulated by thrombin. Emerging antiplatelet therapies that might address these limitations include intravenous P2Y12 antagonists, oral PAR-1 antagonists, and thromboxane-receptor inhibitors. In this Review, we provide an overview of these novel antiplatelet drugs, including newly approved agents and emerging compounds currently under clinical development, and also discuss evolving concepts and unmet needs related to antiplatelet therapy for the treatment of ACS.


Asunto(s)
Síndrome Coronario Agudo/tratamiento farmacológico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores , Receptores de Tromboxanos/antagonistas & inhibidores , Aprobación de Drogas , Descubrimiento de Drogas , Humanos
12.
Exp Parasitol ; 145 Suppl: S78-83, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24703976

RESUMEN

Granulomatous amoebic encephalitis is a rare but serious human disease leading almost always to death. The pathophysiology of amoebic encephalitis is better understood, while events leading to the constitution of brain infection are largely unknown. Traversal of the blood-brain barrier is a key step in amoebae invasion of the central nervous system and facilitated by amoebic extracellular proteases. By using specific inhibitors of protease-activated receptors 1, 2 and 4, here we studied the role of these host receptors in Acanthamoeba castellanii-mediated damage to human brain microvasculature endothelial cells (HBMEC), which constitute the blood-brain barrier. The primary HBMEC were incubated with A. castellanii-conditioned medium in the presence or absence of FR-171113 (selective inhibitor of protease-activated receptor 1), FSLLRY-NH2 (inhibitor of protease-activated receptor 2), and tcY-NH2 (inhibitor of protease-activated receptor 4). The HBMEC monolayer disruptions were assessed by microscopy using Eosin staining, while host cell cytotoxicity was determined by measuring the release of cytoplasmic lactate dehydrogenase. Zymographic assays were performed to determine the effects of inhibitors of protease-activated receptors on the extracellular proteolytic activities of A. castellanii. A. castellanii-conditioned medium produced severe HBMEC monolayer disruptions within 60 min. The selective inhibitors of protease-activated receptors tested did not affect HBMEC monolayer disruptions. On the contrary, pre-treatment of A. castellanii-conditioned medium with phenylmethylsulfonyl fluoride, a serine protease inhibitor, or heating for 10 min at 95°C abolished HBMEC monolayer disruptions. Additionally, inhibitors of protease-activated receptors tested, failed to block A. castellanii-mediated HBMEC cytotoxicity and did not affect extracellular proteolytic activities of A. castellanii. Protease-activated receptors 1, 2 and 4 do not appear to play a role in A. castellanii-mediated dysfunction of HBMEC, which constitute the blood-brain barrier. The role of additional protease-activated receptors in amoebic invasion of the central nervous system is discussed further.


Asunto(s)
Queratitis por Acanthamoeba/parasitología , Acanthamoeba castellanii/fisiología , Células Endoteliales/parasitología , Endotelio Vascular/citología , Microvasos/citología , Receptores Proteinasa-Activados/antagonistas & inhibidores , Queratitis por Acanthamoeba/patología , Acanthamoeba castellanii/aislamiento & purificación , Acanthamoeba castellanii/patogenicidad , Encéfalo/irrigación sanguínea , Encéfalo/citología , Encéfalo/parasitología , Encéfalo/patología , Células Cultivadas , Células Endoteliales/patología , Endotelio Vascular/parasitología , Endotelio Vascular/patología , Humanos , Microvasos/parasitología , Receptor PAR-1/antagonistas & inhibidores , Receptor PAR-2/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores
13.
Med Clin (Barc) ; 143(11): 508-14, 2014 Dec 09.
Artículo en Español | MEDLINE | ID: mdl-24480290

RESUMEN

The dual antiplatelet therapy with acetylsalicylic acid and clopidogrel has been the mainstay of both acute and chronic phase coronary artery disease, reducing importantly the risk of adverse events. Despite a correct compliance, a non-negligible rate of adverse events still happens. New compounds, with improved properties, are now clinically available (such as prasugrel or ticagrelor) or under advanced development. The aim of the present review is the description of these new compounds, particularly prasugrel and ticagrelor.


Asunto(s)
Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Adenosina/efectos adversos , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosina/uso terapéutico , Adenosina Monofosfato/efectos adversos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Ensayos Clínicos Fase III como Asunto , Método Doble Ciego , Disnea/inducido químicamente , Hemorragia/inducido químicamente , Humanos , Estudios Multicéntricos como Asunto , Piperazinas/efectos adversos , Piperazinas/farmacología , Piperazinas/uso terapéutico , Inhibidores de Agregación Plaquetaria/efectos adversos , Inhibidores de Agregación Plaquetaria/clasificación , Inhibidores de Agregación Plaquetaria/farmacología , Clorhidrato de Prasugrel , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptores Proteinasa-Activados/antagonistas & inhibidores , Tiofenos/efectos adversos , Tiofenos/farmacología , Tiofenos/uso terapéutico , Ticagrelor , Resultado del Tratamiento
14.
Thromb Haemost ; 110(2): 295-307, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23739922

RESUMEN

Extravascular activation of the coagulation cascade in the lung is commonly observed in pulmonary fibrosis. Coagulation proteases may exert profibrotic cellular effects via protease-activated receptors (PARs)-1 and -2. Here, we investigated the potential role of two other members of the PAR family, namely PAR-3 and PAR-4, in the pathobiology of lung fibrosis. Elevated expression of PAR-3, but not PAR-4, was detected in the lungs of idiopathic pulmonary fibrosis (IPF) patients and in bleomycin-induced lung fibrosis in mice. Increased PAR-3 expression in fibrotic lungs was mainly attributable to alveolar type II (ATII) cells. Stimulation of primary mouse ATII, MLE15 and A549 cells with thrombin (FIIa) - that may activate PAR-1, PAR-3 and PAR-4 - induced epithelial-mesenchymal transition (EMT), a process that has been suggested to be a possible mechanism underlying the expanded (myo)fibroblast pool in lung fibrosis. EMT was evidenced by morphological alterations, expression changes of epithelial and mesenchymal phenotype markers, and functional changes. Single knockdown of FIIa receptors, PAR-1, PAR-3, or PAR-4, had no major impact on FIIa-induced EMT. Simultaneous depletion of PAR-1 and PAR-3, however, almost completely inhibited this process, whereas only a partial effect on FIIa-mediated EMT was observed when PAR-1 and PAR-4, or PAR-3 and PAR-4 were knocked down. PAR-1 and PAR-3 co-localise within ATII cells with both being predominantely plasma membrane associated. In conclusion, our study indicates that PARs synergise to mediate FIIa-induced EMT and provides first evidence that PAR-3 via its ability to potentiate FIIa-triggered EMT could potentially contribute to the pathogenesis of pulmonary fibrosis.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Fibrosis Pulmonar/etiología , Receptor PAR-1/metabolismo , Receptores Proteinasa-Activados/metabolismo , Receptores de Trombina/metabolismo , Animales , Bleomicina/toxicidad , Diferenciación Celular , Línea Celular , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Ratones , Miofibroblastos/metabolismo , Miofibroblastos/patología , Protrombina/metabolismo , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor PAR-1/antagonistas & inhibidores , Receptor PAR-1/genética , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores Proteinasa-Activados/genética , Receptores de Trombina/antagonistas & inhibidores , Receptores de Trombina/genética , Trombina/metabolismo
16.
Nat Rev Drug Discov ; 11(1): 69-86, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22212680

RESUMEN

Proteinase-activated receptors (PARs), a family of four seven-transmembrane G protein-coupled receptors, act as targets for signalling by various proteolytic enzymes. PARs are characterized by a unique activation mechanism involving the proteolytic unmasking of a tethered ligand that stimulates the receptor. Given the emerging roles of these receptors in cancer as well as in disorders of the cardiovascular, musculoskeletal, gastrointestinal, respiratory and central nervous system, PARs have become attractive targets for the development of novel therapeutics. In this Review we summarize the mechanisms by which PARs modulate cell function and the roles they can have in physiology and diseases. Furthermore, we provide an overview of possible strategies for developing PAR antagonists.


Asunto(s)
Sistemas de Liberación de Medicamentos/tendencias , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/metabolismo , Receptores Proteinasa-Activados/metabolismo , Animales , Sistemas de Liberación de Medicamentos/métodos , Guanidinas/administración & dosificación , Guanidinas/química , Guanidinas/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/metabolismo , Oligopéptidos/administración & dosificación , Oligopéptidos/química , Oligopéptidos/metabolismo , Preparaciones Farmacéuticas/química , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/antagonistas & inhibidores
17.
Br J Pharmacol ; 159(3): 502-17, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20050853

RESUMEN

Platelet inhibitors are the mainstay treatment for patients with vascular diseases. The current 'gold standard' antiplatelet agent clopidogrel has several pharmacological and clinical limitations that have prompted the search for more effective platelet antagonists. The candidates include various blockers of the purinergic P2Y12 receptor such as prasugrel, an oral irreversible thienopyridine; two adenosine triphosphate analogues that bind reversibly to the P2Y12 receptor: ticagrelor (oral) and cangrelor (intravenous); elinogrel, a direct-acting reversible P2Y12 receptor inhibitor (the only antiplatelet compound that can be administered both intravenously and orally); BX 667, an orally active and reversible small-molecule P2Y12 receptor antagonist; SCH 530348, SCH 205831, SCH 602539 and E5555, highly selective and orally active antagonists on the protease-activated receptor 1. A number of drugs also hit new targets: terutroban, an oral, selective and specific inhibitor of the thromboxane receptor; ARC1779, a second-generation, nuclease resistant aptamer which inhibits von Willebrand factor-dependent platelet aggregation; ALX-0081, a bivalent humanized nanobody targeting the GPIb binding site of von Willebrand factor and AJW200, an IgG4 monoclonal antibody of von Willebrand factor. The pharmacology and clinical profiles of new platelet antagonists indicate that they provide more consistent, more rapid and more potent platelet inhibition than agents currently used. Whether these potential advantages will translate into clinical advantages will require additional comparisons in properly powered, randomized, controlled trials.


Asunto(s)
Benzofuranos/farmacología , Plaquetas/efectos de los fármacos , Carbamatos/farmacología , Agregación Plaquetaria/efectos de los fármacos , Piridinas/farmacología , Adenosina/análogos & derivados , Adenosina Monofosfato/análogos & derivados , Ensayos Clínicos como Asunto , Clopidogrel , Humanos , Lactonas , Piperazinas , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacología , Inhibidores de Agregación Plaquetaria/uso terapéutico , Clorhidrato de Prasugrel , Receptor PAR-1 , Receptores Proteinasa-Activados/antagonistas & inhibidores , Tiofenos , Ticagrelor , Ticlopidina/análogos & derivados , Factor de von Willebrand/farmacología
19.
Cardiol Rev ; 16(5): 250-9, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18708826

RESUMEN

Antiplatelet therapy for the prevention and treatment of coronary artery disease (CAD) has undergone dramatic changes and improvements. Aspirin remains the first-line antiplatelet drug for clinical use. Newer platelet inhibitors such as the thienopyridine agents, ticlopidine and clopidogrel, have also been shown to be effective in treating CAD. There have been ongoing efforts to evaluate newer antiplatelet drugs, with the potential to improve clinical efficacy and safety. Some of the more promising antiplatelet agents include new adenosine diphosphate receptor antagonists such as prasugrel, cangrelor, and ticagrelor (AZD6140). In addition, a new thromboxane receptor antagonist, NCX-4016, a newly discovered protease-activated receptor antagonist that targets thrombin-induced platelet aggregation, and anti-von Willebrand factor aptamers show tremendous promise in refining antiplatelet therapy by targeting different receptors and molecules.


Asunto(s)
Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/prevención & control , Inhibidores de Agregación Plaquetaria/uso terapéutico , Humanos , Receptores Proteinasa-Activados/antagonistas & inhibidores , Trombina/antagonistas & inhibidores , Factor de von Willebrand/antagonistas & inhibidores
20.
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