Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Braz. J. Pharm. Sci. (Online) ; 58: e201089, 2022. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1420429

RESUMO

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.


Assuntos
Animais , Feminino , Camundongos , Pneumonia/patologia , Receptor PAR-2/antagonistas & inibidores , Receptores Ativados por Proteinase/antagonistas & inibidores , Remodelação das Vias Aéreas/efeitos dos fármacos
2.
Int J Mol Sci ; 22(7)2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33916310

RESUMO

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.


Assuntos
Quempferóis/uso terapêutico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Embolia Pulmonar/prevenção & controle , Enfisema Pulmonar/prevenção & controle , Receptores Ativados por Proteinase/antagonistas & inibidores , Animais , Fumar Cigarros/efeitos adversos , Avaliação Pré-Clínica de Medicamentos , Quempferóis/farmacologia , Masculino , Camundongos Endogâmicos BALB C , Estresse Oxidativo , Embolia Pulmonar/etiologia
3.
Physiol Rev ; 101(2): 545-567, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33124941

RESUMO

Evolving information has identified disease mechanisms and dysregulation of host biology that might be targeted therapeutically in coronavirus disease 2019 (COVID-19). Thrombosis and coagulopathy, associated with pulmonary injury and inflammation, are emerging clinical features of COVID-19. We present a framework for mechanisms of thrombosis in COVID-19 that initially derive from interaction of SARS-CoV-2 with ACE2, resulting in dysregulation of angiotensin signaling and subsequent inflammation and tissue injury. These responses result in increased signaling by thrombin (proteinase-activated) and purinergic receptors, which promote platelet activation and exert pathological effects on other cell types (e.g., endothelial cells, epithelial cells, and fibroblasts), further enhancing inflammation and injury. Inhibitors of thrombin and purinergic receptors may, thus, have therapeutic effects by blunting platelet-mediated thromboinflammation and dysfunction in other cell types. Such inhibitors include agents (e.g., anti-platelet drugs) approved for other indications, and that could be repurposed to treat, and potentially improve the outcome of, COVID-19 patients. COVID-19, caused by the SARS-CoV-2 virus, drives dysregulation of angiotensin signaling, which, in turn, increases thrombin-mediated and purinergic-mediated activation of platelets and increase in inflammation. This thromboinflammation impacts the lungs and can also have systemic effects. Inhibitors of receptors that drive platelet activation or inhibitors of the coagulation cascade provide opportunities to treat COVID-19 thromboinflammation.


Assuntos
COVID-19/complicações , Inflamação/etiologia , Receptores Ativados por Proteinase/metabolismo , Receptores Purinérgicos/metabolismo , SARS-CoV-2 , Trombose/etiologia , Humanos , Inflamação/tratamento farmacológico , Antagonistas Purinérgicos/farmacologia , Receptores Ativados por Proteinase/antagonistas & inibidores , Receptores Ativados por Proteinase/genética , Receptores Purinérgicos/genética , Trombose/prevenção & controle
4.
Eur J Pharmacol ; 869: 172875, 2020 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-31877279

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Fator Xa/metabolismo , Inflamação/metabolismo , Mitocôndrias/metabolismo , Receptores Ativados por Proteinase/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Dipeptídeos/farmacologia , Células Epiteliais/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Inibidores do Fator Xa/farmacologia , Humanos , Isoxazóis/farmacologia , Mitocôndrias/efeitos dos fármacos , Piridinas/farmacologia , Receptores Ativados por Proteinase/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia
5.
J Leukoc Biol ; 105(4): 729-740, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30690783

RESUMO

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.


Assuntos
Fibrinolisina/metabolismo , Inflamação/patologia , Lipopolissacarídeos/metabolismo , Receptores Ativados por Proteinase/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Peptídeos/farmacologia , Receptores Ativados por Proteinase/agonistas , Receptores Ativados por Proteinase/antagonistas & inibidores , Transdução de Sinais
6.
J Immunol Res ; 2017: 5193572, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28503577

RESUMO

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.


Assuntos
Periodontite/metabolismo , Periodontite/fisiopatologia , Periodonto/metabolismo , Receptor PAR-1/metabolismo , Receptores Ativados por Proteinase/metabolismo , Adesinas Bacterianas/metabolismo , Animais , Células Cultivadas , Cisteína Endopeptidases/metabolismo , Células Epiteliais , Fibroblastos , Regulação da Expressão Gênica , Cisteína Endopeptidases Gingipaínas , Gengiva/citologia , Gengiva/metabolismo , Humanos , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Camundongos , Periodontite/genética , Porphyromonas gingivalis , Receptor PAR-1/agonistas , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-1/genética , Receptores Ativados por Proteinase/agonistas , Receptores Ativados por Proteinase/antagonistas & inibidores , Receptores Ativados por Proteinase/genética
7.
J Thromb Haemost ; 15(4): 597-607, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28079978

RESUMO

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.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fibrose Pulmonar Idiopática/metabolismo , Receptores Ativados por Proteinase/metabolismo , Animais , Anticoagulantes , Bleomicina/química , Fatores de Coagulação Sanguínea/uso terapêutico , Modelos Animais de Doenças , Progressão da Doença , Fibrose/patologia , Humanos , Fibrose Pulmonar Idiopática/mortalidade , Inflamação , Lactonas/uso terapêutico , Camundongos , Inibidores da Agregação Plaquetária/uso terapêutico , Piridinas/uso terapêutico , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-2/antagonistas & inibidores , Receptores Ativados por Proteinase/antagonistas & inibidores
8.
Drugs R D ; 17(1): 65-72, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28063023

RESUMO

INTRODUCTION: Despite the current standard of care, patients with cardiovascular disease remain at a high risk for recurrent events. Inhibition of thrombin-mediated platelet activation through protease-activated receptor-1 antagonism may provide reductions in atherosclerotic disease beyond those achievable with the current standard of care. OBJECTIVE: Our primary objective is to evaluate the clinical literature regarding the role of vorapaxar (Zontivity™) in the reduction of cardiovascular events in patients with a history of myocardial infarction and peripheral artery disease. In particular, we focus on the potential future directions for protease-activating receptor antagonists in the treatment of a broad range of atherosclerotic diseases. DATA SOURCES: A literature search of PubMed and EBSCO was conducted to identify randomized clinical trials from August 2005 to June 2016 using the search terms: 'vorapaxar', 'SCH 530348', 'protease-activated receptor-1 antagonist', and 'Zontivity™'. Bibliographies were searched and additional resources were obtained. RESULTS: Vorapaxar is a first-in-class, protease-activated receptor-1 antagonist. The Thrombin Receptor Antagonist for Clinical Event Reduction (TRACER) trial did not demonstrate a significant reduction in a broad primary composite endpoint. However, the Thrombin-Receptor Antagonist in Secondary Prevention of Atherothrombotic Ischemic Events (TRA 2°P-TIMI 50) trial examined a more traditional composite endpoint and found a significant benefit with vorapaxar. Vorapaxar significantly increased bleeding compared with standard care. Ongoing trials will help define the role of vorapaxar in patients with peripheral arterial disease, patients with diabetes mellitus, and other important subgroups. The use of multivariate modeling may enable the identification of subgroups with maximal benefit and minimal harm from vorapaxar. CONCLUSION: Vorapaxar provides clinicians with a novel mechanism of action to further reduce the burden of ischemic heart disease. Identification of patients with a high ischemic risk and low bleeding risk would enable clinicians to maximize the utility of this unique agent.


Assuntos
Aterosclerose/tratamento farmacológico , Lactonas/farmacologia , Lactonas/uso terapêutico , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Piridinas/farmacologia , Piridinas/uso terapêutico , Receptores Ativados por Proteinase/antagonistas & inibidores , Aterosclerose/metabolismo , Humanos , Comportamento de Redução do Risco
9.
Annu Rev Pharmacol Toxicol ; 57: 349-373, 2017 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-27618736

RESUMO

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.


Assuntos
Descoberta de Drogas/métodos , Receptores Ativados por Proteinase/antagonistas & inibidores , Receptores Ativados por Proteinase/metabolismo , Animais , Descoberta de Drogas/tendências , Humanos , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
10.
Toxicology ; 365: 9-16, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27475285

RESUMO

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.


Assuntos
Colestase/patologia , Cirrose Hepática/patologia , Ativação Plaquetária/efeitos dos fármacos , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Ativados por Proteinase/antagonistas & inibidores , Ticlopidina/análogos & derivados , 1-Naftilisotiocianato , Animais , Doença Hepática Induzida por Substâncias e Drogas/patologia , Colestase/induzido quimicamente , Clopidogrel , Fígado/efeitos dos fármacos , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Ativados por Proteinase/metabolismo , Serotonina/sangue , Ticlopidina/farmacologia , Xenobióticos
11.
Curr Pharm Des ; 22(4): 472-84, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26696253

RESUMO

Protease-activated receptors (PARs) are a unique group of four G-protein coupled receptors. They are widely expressed within the cardiovascular system and the heart. PARs are activated via cleavage by serine proteases. In vitro and in vivo studies showed that the activation of PAR1 and PAR2 plays a crucial role in virus induced inflammatory diseases. The receptors enable cells to recognize pathogen-derived changes in the extracellular environment. An infection with Coxsackie-virus B3 (CVB3) can cause myocarditis. Recent studies have been shown that PAR1 signaling enhanced the antiviral innate immune response via interferon ß (IFNß) and thus limited the virus replication and cardiac damage. In contrast, PAR2 signaling decreased the antiviral innate immune response via IFNß und thus increased the virus replication, which caused severe myocarditis. Along with CVB3 other viruses such as influenza A virus (IAV) and herpes simplex virus (HSV) can induce myocarditis. The role of PAR signaling in IAV infections is contrarily discussed. During HSV infections PARs facilitate the virus infection of the host cell. These studies show that PARs might be interesting drug targets for the treatment of virus infections and inflammatory heart diseases. First studies with PAR agonists, antagonists, and serine protease inhibitors have been conducted in mice. The inhibition of thrombin the main PAR1 activating protease decreased the IFNß response and increased the virus replication in CVB3-induced myocarditis. This indicates that further studies with direct PAR agonists and antagonists are needed to determine whether PARs are useful drug targets for the therapy of virus-induced heart diseases.


Assuntos
Miocardite/tratamento farmacológico , Miocardite/metabolismo , Receptores Ativados por Proteinase/antagonistas & inibidores , Receptores Ativados por Proteinase/metabolismo , Animais , Anticoagulantes/farmacologia , Anticoagulantes/uso terapêutico , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/fisiologia , Miocardite/imunologia , Receptores Ativados por Proteinase/imunologia , Receptores Toll-Like/antagonistas & inibidores , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo
12.
Curr Pharm Des ; 21(30): 4392-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26471318

RESUMO

From the discovery of protease activated receptors (PARs) to the development of first clinically available PAR1 antagonist (vorapaxar) more than two decades of continuous research have passed. There are four different types of PARs named as PAR1, 2, 3 and 4 having a unique mechanism of signaling. These receptors are present in different organs, including the cardiovascular system. Presence of PARs in heart and blood vessels, alteration in the level and activity of the receptors in pathological conditions along with availability of antagonists makes these receptors targetable in several cardiac diseases. Therapeutic benefits of PAR antagonist have been proven in animal model of cardiac diseases such as myocardial infarction, viral myocarditis, atherosclerosis, pulmonary arterial hypertension, etc. PAR signaling plays a vital role in mediating cardiac hypertrophy, inflammation and fibrosis. Apart from having cardiac importance PAR antagonist are also continuously experimented for their beneficial effects in improving insulin resistance in metabolic syndromes. In the present review, we have discussed the functions of individual PARs in the heart and blood vessels along with the expected usefulness of PAR modulators in cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Lactonas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Piridinas/farmacologia , Receptores Ativados por Proteinase/antagonistas & inibidores , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Humanos , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-1/metabolismo , Receptores Ativados por Proteinase/metabolismo
13.
Thromb Res ; 136(6): 1216-23, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26412559

RESUMO

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.


Assuntos
Transfusão de Sangue/métodos , Inibidores da Agregação Plaquetária/química , Agregação Plaquetária/efeitos dos fármacos , Testes de Função Plaquetária , Proteínas Reguladoras de Apoptose/efeitos dos fármacos , Ácido Araquidônico/metabolismo , Aspirina/uso terapêutico , Plaquetas/efeitos dos fármacos , Clopidogrel , Citometria de Fluxo , Hemorragia , Humanos , Lactonas/uso terapêutico , Selectina-P/metabolismo , Fragmentos de Peptídeos/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/uso terapêutico , Transfusão de Plaquetas , Antagonistas do Receptor Purinérgico P2Y/química , Piridinas/uso terapêutico , Receptores Ativados por Proteinase/antagonistas & inibidores , Receptores Purinérgicos P2Y12/metabolismo , Ticlopidina/análogos & derivados , Ticlopidina/uso terapêutico
14.
Handb Exp Pharmacol ; 226: 219-35, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25861783

RESUMO

Protease-activated receptors (PARs) have been implicated in a variety of physiological functions, as well as somatosensation and particularly itch and pain. Considerable attention has focused on PARs following the finding they are upregulated in the skin of atopic dermatitis patients. The present review focuses on recent studies showing that PARs are critically involved in itch and sensitization of itch. PARs are expressed by diverse cell types including primary sensory neurons, keratinocytes, and immune cells and are activated by proteases that expose a tethered ligand. Endogenous proteases are also released from diverse cell types including keratinocytes and immune cells. Exogenous proteases released from certain plants and insects contacting the skin can also induce itch. Increased levels of proteases in the skin contribute to inflammation that is often accompanied by chronic itch which is not predominantly mediated by histamine. The neural pathway signaling itch induced by activation of PARs is distinct from that mediating histamine-induced itch. In addition, there is evidence that PARs play an important role in sensitization of itch signaling under conditions of chronic itch. These recent findings suggest that PARs and other molecules involved in the itch-signaling pathway are good targets to develop novel treatments for most types of chronic itch that are poorly treated with antihistamines.


Assuntos
Prurido/fisiopatologia , Receptores Ativados por Proteinase/fisiologia , Animais , Humanos , Prurido/tratamento farmacológico , Prurido/etiologia , Receptores Ativados por Proteinase/antagonistas & inibidores
15.
Handb Exp Pharmacol ; 227: 239-60, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25846622

RESUMO

Proteinase-activated receptors (PARs) are a family of G protein-coupled receptor that are activated by extracellular cleavage of the receptor in the N-terminal domain. This slicing of the receptor exposes a tethered ligand which binds to a specific docking point on the receptor surface to initiate intracellular signalling. PARs are expressed by numerous tissues in the body, and they are involved in various physiological and pathological processes such as food digestion, tissue remodelling and blood coagulation. This chapter will summarise how serine proteinases activate PARs leading to the development of pain in several chronic pain conditions. The potential of PARs as a drug target for pain relief is also discussed.


Assuntos
Dor/etiologia , Receptores Ativados por Proteinase/fisiologia , Animais , Humanos , Dor/fisiopatologia , Receptores Ativados por Proteinase/antagonistas & inibidores , Serina Proteases/fisiologia , Transdução de Sinais
16.
Arterioscler Thromb Vasc Biol ; 35(3): 500-12, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25633316

RESUMO

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.


Assuntos
Plaquetas/efeitos dos fármacos , Desenho de Fármacos , Terapia de Alvo Molecular , Inibidores da Agregação Plaquetária/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Trombose/tratamento farmacológico , Animais , Plaquetas/metabolismo , Humanos , Inibidores da Agregação Plaquetária/química , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores Ativados por Proteinase/antagonistas & inibidores , Receptores Ativados por Proteinase/metabolismo , Receptores Purinérgicos P2Y12/efeitos dos fármacos , Receptores Purinérgicos P2Y12/metabolismo , Trombose/sangue , Trombose/metabolismo
17.
Nat Rev Cardiol ; 12(1): 30-47, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25286881

RESUMO

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.


Assuntos
Síndrome Coronariana Aguda/tratamento farmacológico , Inibidores da Agregação Plaquetária/uso terapêutico , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Receptores Ativados por Proteinase/antagonistas & inibidores , Receptores de Trombina/antagonistas & inibidores , Receptores de Tromboxanos/antagonistas & inibidores , Aprovação de Drogas , Descoberta de Drogas , Humanos
18.
Exp Parasitol ; 145 Suppl: S78-83, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24703976

RESUMO

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.


Assuntos
Ceratite por Acanthamoeba/parasitologia , Acanthamoeba castellanii/fisiologia , Células Endoteliais/parasitologia , Endotélio Vascular/citologia , Microvasos/citologia , Receptores Ativados por Proteinase/antagonistas & inibidores , Ceratite por Acanthamoeba/patologia , Acanthamoeba castellanii/isolamento & purificação , Acanthamoeba castellanii/patogenicidade , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Encéfalo/parasitologia , Encéfalo/patologia , Células Cultivadas , Células Endoteliais/patologia , Endotélio Vascular/parasitologia , Endotélio Vascular/patologia , Humanos , Microvasos/parasitologia , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-2/antagonistas & inibidores , Receptores de Trombina/antagonistas & inibidores
19.
Int J Mol Sci ; 15(4): 6169-83, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24733067

RESUMO

Protease-activated receptors (PARs) are a family of four G protein-coupled receptors that exhibit increasingly appreciated differences in signaling and regulation both within and between the receptor class. By nature of their proteolytic self-activation mechanism, PARs have unique processes of receptor activation, "ligand" binding, and desensitization/resensitization. These distinctive aspects have presented both challenges and opportunities in the targeting of PARs for therapeutic benefit-the most notable example of which is inhibition of PAR1 on platelets for the prevention of arterial thrombosis. However, more recent studies have uncovered further distinguishing features of PAR-mediated signaling, revealing mechanisms by which identical proteases elicit distinct effects in the same cell, as well as how distinct proteases produce different cellular consequences via the same receptor. Here we review this differential signaling by PARs, highlight how important distinctions between PAR1 and PAR4 are impacting on the progress of a new class of anti-thrombotic drugs, and discuss how these more recent insights into PAR signaling may present further opportunities for manipulating PAR activation and signaling in the development of novel therapies.


Assuntos
Receptores Ativados por Proteinase/metabolismo , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Dimerização , Fibrinolíticos/uso terapêutico , Humanos , Proteína C/metabolismo , Receptores Ativados por Proteinase/agonistas , Receptores Ativados por Proteinase/antagonistas & inibidores , Receptores de Trombina/antagonistas & inibidores , Receptores de Trombina/metabolismo , Sepse/terapia , Transdução de Sinais , Trombose/prevenção & controle , Trombose/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...