Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
1.
Eur Heart J ; 38(1): 38-50, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27071821

RESUMO

AIMS: Atrial fibrillation (AF) produces a hypercoagulable state. Stimulation of protease-activated receptors by coagulation factors provokes pro-fibrotic, pro-hypertrophic, and pro-inflammatory responses in a variety of tissues. We studied the effects of thrombin on atrial fibroblasts and tested the hypothesis that hypercoagulability contributes to the development of a substrate for AF. METHODS AND RESULTS: In isolated rat atrial fibroblasts, thrombin enhanced the phosphorylation of the pro-fibrotic signalling molecules Akt and Erk and increased the expression of transforming growth factor ß1 (2.7-fold) and the pro-inflammatory factor monocyte chemoattractant protein-1 (6.1-fold). Thrombin also increased the incorporation of 3H-proline, suggesting enhanced collagen synthesis by fibroblasts (2.5-fold). All effects could be attenuated by the thrombin inhibitor dabigatran. In transgenic mice with a pro-coagulant phenotype (TMpro/pro), the inducibility of AF episodes lasting >1 s was higher (7 out of 12 vs. 1 out of 10 in wild type) and duration of AF episodes was longer compared with wild type mice (maximum episode duration 42.8 ± 68.4 vs. 0.23 ± 0.39 s). In six goats with persistent AF treated with nadroparin, targeting Factor Xa-mediated thrombin generation, the complexity of the AF substrate was less pronounced than in control animals (LA maximal activation time differences 23.3 ± 3.1 ms in control vs. 15.7 ± 2.1 ms in nadroparin, P < 0.05). In the treated animals, AF-induced α-smooth muscle actin expression was lower and endomysial fibrosis was less pronounced. CONCLUSION: The hypercoagulable state during AF causes pro-fibrotic and pro-inflammatory responses in adult atrial fibroblasts. Hypercoagulability promotes the development of a substrate for AF in transgenic mice and in goats with persistent AF. In AF goats, nadroparin attenuates atrial fibrosis and the complexity of the AF substrate. Inhibition of coagulation may not only prevent strokes but also inhibit the development of a substrate for AF.


Assuntos
Fibrilação Atrial/etiologia , Receptores de Trombina/efeitos dos fármacos , Trombina/farmacologia , Trombofilia/fisiopatologia , Análise de Variância , Animais , Antitrombinas/farmacologia , Proliferação de Células/efeitos dos fármacos , Dabigatrana/farmacologia , Feminino , Fibrinolíticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibrose/etiologia , Cabras , Átrios do Coração/patologia , Indazóis/farmacologia , Camundongos Transgênicos , Nadroparina/farmacologia , Peptídeo Hidrolases/efeitos dos fármacos , Pirróis/farmacocinética , Quinazolinas/farmacocinética , Ratos , Ureia/análogos & derivados , Ureia/farmacologia
2.
Arterioscler Thromb Vasc Biol ; 36(5): 952-60, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26966273

RESUMO

OBJECTIVE: Protease-activated receptor 4 (PAR4) is a key regulator of platelet reactivity and is encoded by F2RL3, which has abundant rare missense variants. We aimed to provide proof of principle that rare F2LR3 variants potentially affect platelet reactivity and responsiveness to PAR1 antagonist drugs and to explore underlying molecular mechanisms. APPROACH AND RESULTS: We identified 6 rare F2RL3 missense variants in 236 cardiac patients, of which the variant causing a tyrosine 157 to cysteine substitution (Y157C) was predicted computationally to have the greatest effect on PAR4 structure. Y157C platelets from 3 cases showed reduced responses to PAR4-activating peptide and to α-thrombin compared with controls, but no reduction in responses to PAR1-activating peptide. Pretreatment with the PAR1 antagonist vorapaxar caused lower residual α-thrombin responses in Y157C platelets than in controls, indicating greater platelet inhibition. HEK293 cells transfected with a PAR4 Y157C expression construct had reduced PAR4 functional responses, unchanged total PAR4 expression but reduced surface expression. PAR4 Y157C was partially retained in the endoplasmic reticulum and displayed an expression pattern consistent with defective N-glycosylation. Mutagenesis of Y322, which is the putative hydrogen bond partner of Y157, also reduced PAR4 surface expression in HEK293 cells. CONCLUSIONS: Reduced PAR4 responses associated with Y157C result from aberrant anterograde surface receptor trafficking, in part, because of disrupted intramolecular hydrogen bonding. Characterization of PAR4 Y157C establishes that rare F2RL3 variants have the potential to markedly alter platelet PAR4 reactivity particularly after exposure to therapeutic PAR1 antagonists.


Assuntos
Plaquetas/metabolismo , Ativação Plaquetária , Receptores de Trombina/metabolismo , Idoso , Plaquetas/efeitos dos fármacos , Estudos de Casos e Controles , Simulação por Computador , Relação Dose-Resposta a Droga , Retículo Endoplasmático/metabolismo , Inglaterra , Feminino , Genótipo , Glicosilação , Células HEK293 , Humanos , Ligação de Hidrogênio , Lactonas/farmacologia , Masculino , Modelos Moleculares , Mutação de Sentido Incorreto , Peptídeos/farmacologia , Fenótipo , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária , Inibidores da Agregação Plaquetária/farmacologia , Polimorfismo de Nucleotídeo Único , Conformação Proteica , Transporte Proteico , Piridinas/farmacologia , Receptor PAR-1/efeitos dos fármacos , Receptor PAR-1/metabolismo , Receptores de Trombina/química , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/genética , Relação Estrutura-Atividade , Trombina/farmacologia , Transfecção
3.
Neurochem Res ; 40(11): 2220-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26342829

RESUMO

Thrombin's role in the nervous system is not well understood. Under conditions of blood-brain barrier compromise (e.g., neurosurgery or stroke), thrombin can result in neuroapoptosis and the formation of glial scars. Despite this, preconditioning with thrombin has been found to be neuroprotective in models of cerebral ischemia and intracerebral hemorrhage. We investigated the effects of physiologically relevant concentrations of thrombin on cortical neurons using two culture-based assays. We examined thrombin's effect on neurites by quantitative analysis of fluorescently labeled neurons. To characterize thrombin's effects on neuron survival, we spectrophotometrically measured changes in enzymatic activity. Using receptor agonists and thrombin inhibitors, we separately examined the role of thrombin and its receptor in neuroprotection. We found that low concentrations of thrombin (1 nM) enhances neurite growth and branching, neuron viability, and protects against excitotoxic damage. In contrast, higher concentrations of thrombin (100 nM) are potentially detrimental to neuronal health as evidenced by inhibition of neurite growth. Lower concentrations of thrombin resulted in equivalent neuroprotection as the antifibrinolytic, aprotinin, and the direct thrombin inhibitor, argatroban. Interestingly, exogenous application of the species-specific thrombin inhibitor, antithrombin III, was detrimental to neuronal health; suggesting that some endogenous thrombin is necessary for optimal neuron health in our culture system. Activation of the thrombin receptor, protease-activated receptor-1 (PAR-1), via micromolar concentrations of the thrombin receptor agonist peptide, TRAP, did not adversely affect neuronal viability. An optimal concentration of thrombin exists to enhance neuronal health. Neurotoxic effects of thrombin do not involve activation of PAR receptors and thus separate pharmacologic manipulation of thrombin's receptor in the setting of direct thrombin inhibitors could be a potential neuroprotective strategy.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Trombina/farmacologia , Animais , Antitrombina III/farmacologia , Aprotinina/farmacologia , Arginina/análogos & derivados , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Neuritos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Ácidos Pipecólicos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor PAR-1/agonistas , Receptores de Trombina/efeitos dos fármacos , Sulfonamidas , Trombina/antagonistas & inibidores
4.
J Thromb Haemost ; 12(8): 1331-41, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24888424

RESUMO

BACKGROUND: Human platelet activation and aggregation is a complex process. To date, many therapies have been developed targeting proteins that mediate this process to prevent unwanted activation. However, the current standard of care for acute coronary syndromes still has limitations, including bleeding risk. OBJECTIVE: To evaluate the protease-activated receptor 4 (PAR4) anionic cluster as a viable antiplatelet target by using a polyclonal antibody (CAN12). METHODS: We used western blotting, aggregation and secretion ex vivo to evaluate the ability of CAN12 to interact with PAR4 and inhibit platelet activation. The effects of CAN12 in vivo were evaluated with the Rose Bengal arterial thrombosis model and two models of hemostasis. RESULTS: CAN12 was able to interact with human PAR4 and delay PAR4 cleavage. In addition, CAN12 inhibited thrombin-induced human platelet aggregation and secretion in a dose-dependent manner. The specificity of CAN12 was agonist-dependent. In vivo, we determined that CAN12 was able to inhibit arterial thrombosis, and, using two independent methods, we found that CAN12 did not influence hemostasis. CONCLUSION: Targeting the extracellular anionic cluster on PAR4 is a viable novel strategy as an antiplatelet therapy.


Assuntos
Anticorpos/farmacologia , Hemostasia/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Receptores de Trombina/efeitos dos fármacos , Trombose/prevenção & controle , Animais , Western Blotting , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ativação Plaquetária/efeitos dos fármacos
5.
Arterioscler Thromb Vasc Biol ; 32(11): 2609-15, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22995523

RESUMO

OBJECTIVE: Statins decrease mortality in patients with vascular disorders, and evidence for the pleiotropic effects of statins is accumulating. Statins enhance endothelial NO synthase (eNOS) expression, thereby attenuating platelet activation and thrombus formation. Our goal was to determine whether statins have eNOS-independent effects on platelet activation. METHODS AND RESULTS: Wild-type and eNOS-deficient mice were given a 14-day course of oral atorvastatin, and platelet activation was evaluated in vitro and in vivo. Whereas in wild-type mice atorvastatin inhibited platelet activation in vitro in response to numerous agonists, in eNOS-deficient mice, atorvastatin inhibited only thrombin-induced and protease-activated receptor 4 agonist peptide-induced platelet activation. Consistent with an eNOS-independent effect, atorvastatin inhibited platelet activation in vivo in both wild-type and eNOS-deficient mice. CONCLUSIONS: Atorvastatin inhibits platelet activation via eNOS-dependent and eNOS-independent mechanisms with the latter restricted to protease-activated receptor 4-induced activation downstream to the receptor.


Assuntos
Plaquetas/efeitos dos fármacos , Ácidos Heptanoicos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Óxido Nítrico Sintase Tipo III/deficiência , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Pirróis/farmacologia , Administração Oral , Animais , Atorvastatina , Plaquetas/metabolismo , GMP Cíclico/metabolismo , Ácidos Heptanoicos/administração & dosagem , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/genética , Selectina-P/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/administração & dosagem , Testes de Função Plaquetária , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Pirróis/administração & dosagem , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trombina/metabolismo , Fatores de Tempo
6.
Adv Cardiol ; 47: 87-99, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22906905

RESUMO

The unifying basis of acute coronary syndrome (ACS) is the complication of a vulnerable coronary plaque, an event primarily mediated by platelet activation. Three major pathways are predominantly involved in this process: thromboxane A(2) via the thromboxane A(2) receptor, adenosine diphosphate via the P2Y(12) receptor, and thrombin via the protease-activated receptor (PAR)-1, with the latter being the most potent platelet activator. Despite the effective inhibition of the first two pathways with aspirin and an expanding family of P2Y(12) inhibitors, respectively, the recurrence of ischemic events in patients with ACS remains high. There is also a growing concern regarding the safety profile in terms of bleeding with more powerful antiplatelet agents, which has tempered expectations of newly developed compounds. PAR-1 inhibitors are a novel class of antiplatelet agents that inhibit thrombin-mediated platelet activation. Preliminary data indicate that these compounds have the potential to improve ischemic prognosis without increasing the bleeding risk. In this chapter we will discuss the rationale for developing this novel class of antiplatelet agents and specifically, the two compounds in most advanced clinical development, vorapaxar and atopaxar.


Assuntos
Síndrome Coronariana Aguda/tratamento farmacológico , Lactonas/uso terapêutico , Piridinas/uso terapêutico , Receptor PAR-1/antagonistas & inibidores , Síndrome Coronariana Aguda/complicações , Síndrome Coronariana Aguda/fisiopatologia , Animais , Humanos , Iminas/administração & dosagem , Iminas/química , Iminas/farmacologia , Iminas/uso terapêutico , Lactonas/administração & dosagem , Lactonas/química , Lactonas/farmacologia , Isquemia Miocárdica/complicações , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Piridinas/administração & dosagem , Piridinas/química , Piridinas/farmacologia , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/fisiologia
7.
Pharmacogenet Genomics ; 22(1): 43-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22082654

RESUMO

BACKGROUND: Platelet aggregation varies among individuals; and genetic factors may alter platelet activation through G-protein-coupled receptors, thus influencing results of point-of-care platelet aggregometry in whole blood. We tested the hypothesis that the C825T polymorphism of the gene GNB3 encoding the G-protein ß-3 subunit and the platelet GPIIIa Pl(A1)/(A2) polymorphism of the glycoprotein IIIa influence platelet aggregation. METHODS: Evoked [thrombin receptor activating peptide (TRAP), ADP, TXA(2) agonist U46619, epinephrine, and collagen] platelet aggregation in whole blood was measured using impedance aggregometry (Multiplate) in 143 healthy individuals (age: 40.2 years ±11.7 SD). Genotypes were determined using pyrosequencing and restriction analysis. Data were analyzed by linear one-way analysis of variance and Student's t-test, linear and multiple regression, and the χ(2)-test, as appropriate. RESULTS: Homozygous carriers of the GNB3 825C-allele showed significantly (P≤0.022) increased maximum aggregation for EC(75) dosages compared with CT and TT genotypes [e.g. ADP: CC 150±36 vs. TT 126±33 aggregation unit (AU); thrombin receptor activating peptide: CC 175±46 vs. TT 150±38 AU; U46619: CC 164±33 vs. 149±32 AU; epinephrine: CC 66±41 vs. TT 48±34 AU]. In contrast, genotypes of glycoprotein IIb/IIIa PI(A)-polymorphism had no effect. Regression analysis revealed the GNB3 C825T polymorphism as an independent factor for enhanced platelet aggregation, besides factors such as female sex and blood cell values. CONCLUSION: In human whole blood, the GNB3 825CC genotype is associated with enhanced platelet aggregation.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/genética , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/genética , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Adolescente , Adulto , Alelos , Colágeno/farmacologia , Epinefrina/farmacologia , Genótipo , Proteínas Heterotriméricas de Ligação ao GTP/sangue , Heterozigoto , Humanos , Integrina beta3/sangue , Integrina beta3/genética , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/farmacologia , Polimorfismo de Nucleotídeo Único/genética , Receptores Purinérgicos P2Y12/efeitos dos fármacos , Receptores Purinérgicos P2Y12/metabolismo , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/metabolismo
8.
J Thromb Haemost ; 9(8): 1599-607, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21649850

RESUMO

BACKGROUND: Cytosolic calcium concentration is a critical regulator of platelet activation, and so platelet Ca(2+) signaling must be tightly controlled. Thrombin-induced Ca(2+) signaling is enhanced by inhibitors of protein kinase C (PKC), suggesting that PKC negatively regulates the Ca(2+) signal, although the mechanisms by which this occurs and its physiological relevance are still unclear. OBJECTIVES: To investigate the mechanisms by which PKC inhibitors enhance thrombin-induced Ca(2+) signaling, and to determine the importance of this pathway in platelet activation. METHODS: Cytosolic Ca(2+) signaling was monitored in fura-2-loaded human platelets. Phosphatidylserine (PS) exposure, a marker of platelet procoagulant activity, was measured by annexin V binding and flow cytometry. RESULTS: PKC inhibition by bisindolylmaleimide-I (BIM-I) enhanced α-thrombin-induced Ca(2+) signaling in a concentration-dependent manner. PAR1 signaling, activated by SFLLRN, was enhanced much more strongly than PAR4, activated by AYPGKF or γ-thrombin, which is a potent PAR4 agonist but a poor activator of PAR1. BIM-I had little effect on α-thrombin-induced signaling following treatment with the PAR1 antagonist, SCH-79797. BIM-I enhanced Ca(2+) release from intracellular stores and Ca(2+) entry, as assessed by Mn(2+) quench. However, the plasma membrane Ca(2+) ATPase inhibitor, 5(6)-carboxyeosin, did not prevent the effect of BIM-I. PKC inhibition strongly enhanced α-thrombin-induced PS exposure, which was reversed by blockade of PAR1. CONCLUSIONS: Together, these data show that when PAR1 is stimulated, PKC negatively regulates Ca(2+) release and Ca(2+) entry, which leads to reduced platelet PS exposure.


Assuntos
Plaquetas/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Indóis/farmacologia , Maleimidas/farmacologia , Fosfatidilserinas/metabolismo , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Receptor PAR-1/efeitos dos fármacos , Receptores de Trombina/efeitos dos fármacos , Anexina A5/metabolismo , Plaquetas/enzimologia , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Proteína Quinase C/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Trombina/metabolismo , Fatores de Tempo
9.
Am J Hematol ; 85(8): 584-92, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20658588

RESUMO

In the Bernard-Soulier syndrome (BSS), the giant platelets are said to have increased phosphatidylserine (PS) surface exposure in the resting state and shortened survival in the circulation. When normal platelets are activated, they undergo many biochemical and morphological changes, some of which are apoptotic. Herein, we investigated apoptotic-like events in BSS platelets upon activation, specifically, PS exposure, microparticle (MP) formation, cell shrinkage, and loss of mitochondrial inner membrane potential (DeltaPsi(m)). Platelets from two unrelated BSS patients were examined in whole blood; agonists used were collagen, thrombin, PAR1- or PAR4-activating peptides (APs), or combinations of collagen with thrombin, and the PAR-APs. Flow cytometry was used to measure PS exposure (annexin A5 binding), platelet-derived MPs (forward scatter; events <0.75 microm size), and DeltaPsi(m) (TMRM fluorescence). PS exposure was increased on resting and activated BSS platelets, and this was independent of the platelet size. MP formation by BSS platelets was generally enhanced. Cell shrinkage occurred on activation to form smaller, PS-exposing platelets in BSS and controls. A proportion of PS-exposing BSS and control platelets exhibited DeltaPsi(m) loss, but unlike controls, there was also loss of DeltaPsi(m) in the BSS platelets not exposing PS. Thus, BSS platelets undergo apoptotic-like events upon activation, with PS exposure and MP formation being enhanced. These events may play a role in the shortened survival in BSS, as well as affecting thrombin generation.


Assuntos
Apoptose , Síndrome de Bernard-Soulier/sangue , Plaquetas/patologia , Lipídeos de Membrana/sangue , Fosfatidilserinas/sangue , Adolescente , Plaquetas/química , Plaquetas/efeitos dos fármacos , Tamanho Celular , Criança , Colágeno/farmacologia , Humanos , Masculino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Oligopeptídeos/biossíntese , Oligopeptídeos/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Contagem de Plaquetas , Complexo Glicoproteico GPIb-IX de Plaquetas/genética , Receptor PAR-1/efeitos dos fármacos , Receptores de Trombina/biossíntese , Receptores de Trombina/efeitos dos fármacos
10.
J Physiol ; 588(Pt 7): 1171-7, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20142268

RESUMO

The nature of protease-activated receptors (PARs) capable of activating respiratory vagal C-fibres in the mouse was investigated. Infusing thrombin or trypsin via the trachea strongly activated vagal lung C-fibres with action potential discharge, recorded with the extracellular electrode positioned in the vagal sensory ganglion. The intensity of activation was similar to that observed with the TRPV1 agonist, capsaicin. This was mimicked by the PAR1-activating peptide TFLLR-NH(2), whereas the PAR2-activating peptide SLIGRL-NH(2) was without effect. Patch clamp recording on cell bodies of capsaicin-sensitive neurons retrogradely labelled from the lungs revealed that TFLLR-NH(2) consistently evokes a large inward current. RT-PCR revealed all four PARs were expressed in the vagal ganglia. However, when RT-PCR was carried out on individual neurons retrogradely labelled from the lungs it was noted that TRPV1-positive neurons (presumed C-fibre neurons) expressed PAR1 and PAR3, whereas PAR2 and PAR4 were rarely expressed. The C-fibres in mouse lungs isolated from PAR1(-/-) animals responded normally to capsaicin, but failed to respond to trypsin, thrombin, or TFLLR-NH(2). These data show that the PAR most relevant for evoking action potential discharge in vagal C-fibres in mouse lungs is PAR1, and that this is a direct neuronal effect.


Assuntos
Pulmão/inervação , Fibras Nervosas Amielínicas/fisiologia , Receptor PAR-1/fisiologia , Trombina/fisiologia , Tripsina/fisiologia , Nervo Vago/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Vias Aferentes/fisiologia , Animais , Capsaicina/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibras Nervosas Amielínicas/efeitos dos fármacos , Oligopeptídeos/fisiologia , Receptor PAR-1/agonistas , Receptor PAR-1/genética , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/fisiologia , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/fisiologia , Trombina/farmacologia , Tripsina/farmacologia , Nervo Vago/efeitos dos fármacos
11.
Br J Pharmacol ; 158(6): 1548-56, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19814731

RESUMO

BACKGROUND AND PURPOSE: The modulation by flavonoids of platelet responses induced by thrombin has been little investigated, and the antiplatelet activity, as well as possible inhibitory mechanisms of these compounds on thrombin signalling, has not yet been elucidated. We explored whether flavonoids affect platelet signalling pathways triggered by thrombin and by the selective activation of its protease-activated receptors (PARs) 1 and 4, and analysed the antagonism of these polyphenols at thrombin receptors. EXPERIMENTAL APPROACH: We investigated the effect of a range of polyphenolic compounds on platelet aggregation, 5-HT secretion, intracellular calcium mobilization, protein kinase activity and tyrosine phosphorylation, triggered by thrombin and PAR agonist peptides (PAR-APs). The ability of these flavonoids to bind to thrombin receptors was investigated by competitive radioligand binding assays using (125)I-thrombin. KEY RESULTS: Quercetin, apigenin and genistein impaired platelet aggregation, as well as 5-HT release and calcium mobilization, induced by thrombin and PAR-APs. Quercetin and apigenin were inhibitors of protein kinases, but genistein exhibited a minimal ability to suppress platelet phosphorylation. Binding assays did not establish any kind of interaction between thrombin receptors and any of the flavonoids tested. CONCLUSIONS AND IMPLICATIONS: Quercetin, apigenin and genistein did not inhibit thrombin responses by interacting with thrombin receptors, but by interfering with intracellular signalling. While inhibition by genistein may be a consequence of affecting calcium mobilization, subsequent platelet secretion and aggregation, for quercetin and apigenin, inhibition of kinase activation may also be involved in the impairment of platelet responses.


Assuntos
Apigenina/farmacologia , Genisteína/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Quercetina/farmacologia , Cálcio/metabolismo , Flavonoides/farmacologia , Humanos , Inibidores da Agregação Plaquetária/farmacologia , Ensaio Radioligante , Receptor PAR-1/efeitos dos fármacos , Receptor PAR-1/metabolismo , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/metabolismo , Serotonina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trombina/metabolismo
12.
Future Cardiol ; 5(5): 435-42, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19715408

RESUMO

SCH 530348, a synthetic tricyclic 3-phenylpyridine, is an orally active fourth generation himbacine-based antagonist of the protease-activated receptor (PAR)-1, the primary receptor for thrombin on platelets in humans. SCH 530348 is the first in a new class of compounds that inhibit thrombin-mediated platelet aggregation without affecting the enzymatic activity of thrombin on fibrinogen. Preclinical and initial clinical studies have demonstrated this compound to be a highly potent inhibitor of thrombin-induced platelet activation, to have excellent oral bioavailability and to have a favorable safety profile. These data suggest that this compound has the potential to reduce the risk of ischemic events without significantly increasing the rate of bleeding. Two large Phase III clinical outcome trials are currently underway to evaluate the safety and efficacy of SCH 530348 for the management of acute coronary syndromes and the secondary prevention of atherothrombotic events.


Assuntos
Síndrome Coronariana Aguda/tratamento farmacológico , Lactonas/uso terapêutico , Agregação Plaquetária/efeitos dos fármacos , Piridinas/uso terapêutico , Receptor PAR-1/antagonistas & inibidores , Receptores de Trombina/antagonistas & inibidores , Trombose Coronária/tratamento farmacológico , Humanos , Lactonas/química , Lactonas/farmacocinética , Piridinas/química , Piridinas/farmacocinética , Receptor PAR-1/efeitos dos fármacos , Receptores de Trombina/efeitos dos fármacos , Tromboembolia/tratamento farmacológico
13.
Future Cardiol ; 5(5): 479-93, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19715412

RESUMO

The present review focuses on the roles of thromboxane A2 (TxA2) in arterial thrombosis, atherogenesis, vascular stent-related ischemic events and renal proteinuria. Particular emphasis is laid on therapeutic interventions targeting the TxA2 (TP) receptors and TxA2 synthase (TS), including dual TP-receptor antagonists and TS inhibitors. Their significant inhibitory efficacies on arterial thrombogenesis, atherogenesis, restenosis after stent placement, vasoconstriction and proteinuria indicate novel and improved treatments for cardiovascular and selected renal diseases. New therapeutic interventions of the TxA2 pathway may also be beneficial for patients with poor biological antiplatelet drug response, for example, to aspirin and/or clopidogrel. These new TP/TS agents offer novel improved treatments to efficiently and simultaneously interfere with thrombogenesis and atherogenesis, and to enlarge the existing panel of platelet inhibitors for efficient prophylaxis and treatment of arterial thrombosis and renal proteinuria.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Nefropatias/tratamento farmacológico , Receptores de Trombina/efeitos dos fármacos , Tromboxano A2/metabolismo , Tromboxano-A Sintase/antagonistas & inibidores , Aspirina/uso terapêutico , Doenças Cardiovasculares/enzimologia , Clopidogrel , Doença da Artéria Coronariana/tratamento farmacológico , Doença da Artéria Coronariana/enzimologia , Reestenose Coronária/tratamento farmacológico , Humanos , Nefropatias/enzimologia , Inibidores da Agregação Plaquetária/uso terapêutico , Proteinúria/tratamento farmacológico , Proteinúria/enzimologia , Receptores de Trombina/antagonistas & inibidores , Trombose/tratamento farmacológico , Trombose/enzimologia , Tromboxano-A Sintase/efeitos dos fármacos , Tromboxano-A Sintase/metabolismo , Ticlopidina/análogos & derivados , Ticlopidina/uso terapêutico
14.
Int J Cancer ; 124(7): 1517-25, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19058300

RESUMO

Thrombin is now recognized as an important factor in many cancers. Here, we examined the expression and role of the recently discovered thrombin receptor PAR4, in human colon cancer cells. PAR4 mRNA was found in 10 out of 14 (71%) human colon cancer cell lines tested but not in epithelial cells isolated from normal human colon. This finding is in line with immunostaining results of PAR4 in human colon tumors and its absence in normal human colonic mucosa. Investigation of the functional significance of the aberrant expression of PAR4 in colon cancer cells revealed (i) a prompt increase in intracellular calcium concentration on challenge with PAR4-specific agonist AP4 (100 microM) and (ii) marked mitogenic response (2.5-fold increase in cell number) in a dose-dependent manner on treatment with AP4 (0.1-300 microM). Analysis of the signaling pathways downstream of PAR4 activation in HT29 cells showed (i) a sustained phosphorylation of extracellular signal-related kinase 1/2 (ERK1/2) and (ii) the involvement of epidermal growth factor receptor B-2 (ErbB-2) but not of epidermal growth factor receptor in PAR4-induced mitogen-activated protein kinase activation. Tyrphostin AG1478, the ErbB inhibitor, reversed the action of AP4 on ERK1/2 and ErbB-2 phosphorylation and HT29 cell growth. Finally, the Src inhibitor PP2 abrogated ErbB-2 and ERK phosphorylation and HT29 cell proliferation, suggesting the essential role of Src activity in PAR4-induced phosphorylation of ErbB-2. These data highlight the role of PAR4 as a new important player in the control of colon tumors and underline the critical role of ErbB-2 transactivation.


Assuntos
Neoplasias do Colo/metabolismo , Receptor ErbB-2/metabolismo , Receptores de Trombina/metabolismo , Transdução de Sinais/fisiologia , Quinases da Família src/metabolismo , Western Blotting , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/patologia , Inibidores Enzimáticos/farmacologia , Imunofluorescência , Células HT29 , Humanos , Imuno-Histoquímica , Imunoprecipitação , RNA Mensageiro/análise , Receptor ErbB-2/efeitos dos fármacos , Receptores de Trombina/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional , Quinases da Família src/efeitos dos fármacos
15.
J Vasc Res ; 46(2): 98-102, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18607113

RESUMO

A synthetic peptide representing the receptor-binding domain of human thrombin (TP508) promotes angiogenesis and accelerates wound healing in animal models. However, the mechanisms underlying the therapeutic effects of TP508 have not been clearly defined. In this study, we set out to determine whether TP508 could stimulate stem cell proliferation. Adipose tissue-derived stem cells (ASCs) were incubated with TP508 (5 microg/ml) and cell proliferation was determined by bromodeoxyuridine (BrdU) incorporation. Our data showed that TP508 treatment significantly stimulated BrdU incorporation in ASCs (p < 0.01). The increased BrdU incorporation induced by TP508 was abolished by the PI3 kinase (PI3K) inhibitor LY294002 at 50 microM. Western blot analysis of ASCs revealed increased phosphorylation of Akt in response to TP508 when compared to unstimulated controls. These results indicate that TP508 exerts proliferative effects on ASCs via the PI3K/Akt pathway.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Gordura Subcutânea/efeitos dos fármacos , Trombina/farmacologia , Bromodesoxiuridina/metabolismo , Células Cultivadas , Cromonas/farmacologia , Humanos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/metabolismo , Células-Tronco/enzimologia , Gordura Subcutânea/enzimologia , Cicatrização/efeitos dos fármacos
16.
Blood ; 112(5): 1696-703, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18544684

RESUMO

Second messenger-mediated inside-out activation of integrin alphaIIbbeta3 is a key step in platelet aggregation. We recently showed strongly impaired but not absent alphaIIbbeta3-mediated aggregation of CalDAG-GEFI-deficient platelets activated with various agonists. Here we further evaluated the roles of CalDAG-GEFI and protein kinase C (PKC) for alphaIIbbeta3 activation in platelets activated with a PAR4 receptor-specific agonist, GYPGKF (PAR4p). Compared with wild-type controls, platelets treated with the PKC inhibitor Ro31-8220 or CalDAG-GEFI-deficient platelets showed a marked defect in aggregation at low (< 1mM PAR4p) but not high PAR4p concentrations. Blocking of PKC function in CalDAG-GEFI-deficient platelets, how-ever, strongly decreased aggregation at all PAR4p concentrations, demonstrating that CalDAG-GEFI and PKC represent separate, but synergizing, pathways important for alphaIIbbeta3 activation. PAR4p-induced aggregation in the absence of CalDAG-GEFI required cosignaling through the Galphai-coupled receptor for ADP, P2Y12. Independent roles for CalDAG-GEFI and PKC/Galphai signaling were also observed for PAR4p-induced activation of the small GTPase Rap1, with CalDAG-GEFI mediating the rapid but reversible activation of this small GTPase. In summary, our study identifies CalDAG-GEFI and PKC as independent pathways leading to Rap1 and alphaIIbbeta3 activation in mouse platelets activated through the PAR4 receptor.


Assuntos
Plaquetas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/sangue , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteína Quinase C/sangue , Difosfato de Adenosina/sangue , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Degranulação Celular/efeitos dos fármacos , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/sangue , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Cinética , Camundongos , Camundongos Knockout , Modelos Biológicos , Oligopeptídeos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Proteína Quinase C/antagonistas & inibidores , Receptores de Trombina/sangue , Receptores de Trombina/efeitos dos fármacos , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/sangue
17.
J Am Coll Cardiol ; 52(1): 33-9, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18582632

RESUMO

OBJECTIVES: We sought to test the hypothesis that cigarette smoking adversely alters protease-activated receptor type 1 (PAR-1)-mediated vascular effects in vivo in humans. BACKGROUND: Distinct from its role in the coagulation cascade, thrombin exerts its major cellular and cardiovascular actions via PAR-1. The activation of PAR-1 causes endothelium-dependent arterial vasodilation and the release of endogenous fibrinolytic factors. METHODS: Forearm blood flow was measured with venous occlusion plethysmography in 12 cigarette smokers and 12 age- and gender-matched nonsmokers during intrabrachial infusions of PAR-1-activating-peptide (SFLLRN; 5 to 50 nmol/min), bradykinin (100 to 1,000 pmol/min), and sodium nitroprusside (2 to 8 mug/min). Plasma tissue plasminogen activator (t-PA) and plasminogen-activator inhibitor 1 antigen and activity concentrations were measured throughout the experiment. RESULTS: All agonists caused dose-dependent increases in forearm blood flow (p < 0.0001 for all). Although bradykinin and sodium nitroprusside caused similar vasodilation, SFLLRN-induced vasodilation was attenuated in smokers (p = 0.04). Smokers had modest reductions in bradykinin-induced active t-PA release (reduced by 37%, p = 0.03) and had a marked impairment of SFLLRN-induced t-PA antigen (p = 0.02) and activity (p = 0.006) release, with a 96% reduction in overall net t-PA antigen release. The use of SFLLRN also caused similar (p = NS) increases in inactive plasminogen-activator inhibitor 1 in both smokers and nonsmokers (p

Assuntos
Fibrinólise , Receptor PAR-1/metabolismo , Fumar/efeitos adversos , Fumar/metabolismo , Trombose/etiologia , Ativador de Plasminogênio Tecidual/metabolismo , Vasodilatação , Adulto , Biomarcadores , Bradicinina/uso terapêutico , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Feminino , Fibrinólise/efeitos dos fármacos , Antebraço/irrigação sanguínea , Hemodinâmica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Nitroprussiato/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Inibidor 1 de Ativador de Plasminogênio/análise , Inibidor 1 de Ativador de Plasminogênio/imunologia , Pletismografia , Receptores de Trombina/efeitos dos fármacos , Projetos de Pesquisa , Fatores de Risco , Fumar/sangue , Fumar/fisiopatologia , Trombose/sangue , Trombose/metabolismo , Trombose/fisiopatologia , Trombose/prevenção & controle , Ativador de Plasminogênio Tecidual/sangue , Vasodilatação/efeitos dos fármacos , Vasodilatadores/uso terapêutico
18.
J Neurochem ; 105(3): 750-62, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18179476

RESUMO

We have performed intrastriatal injection of thrombin and searched for distant effects in the cell body region. In striatum, thrombin produced a slight loss of striatal neurons as demonstrated by neural nuclei immunostaining - a non-specific neuronal marker - and the expression of glutamic acid decarboxylase 67 mRNA, a specific marker for striatal GABAergic interneurons, the most abundant phenotype in this brain area. Interestingly, striatal neuropil contained many boutons immunostained for synaptic vesicle protein 2 and synaptophysin which colocalize with tyrosine hydroxylase (TH), suggesting a degenerative process with pre-synaptic accumulation of synaptic vesicles. When we studied the effects on substantia nigra, we found the disappearance of dopaminergic neurons, shown by loss of TH immunoreactivity, loss of expression of TH and dopamine transporter mRNAs, and disappearance of FluoroGold-labelled nigral neurons. The degeneration of substantia nigra dopaminergic neurons was produced through up-regulation of cFos mRNA, apoptosis and accumulation of alpha-synuclein shown by colocalization experiments. Thrombin effects could be mediated by protease-activated receptor 4 activation, as protease-activated receptor 4-activating peptide mimicked thrombin effects. Our results point out the possible relationship between synapse elimination and retrograde degeneration in the nigral dopaminergic system.


Assuntos
Apoptose/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Degeneração Retrógrada/induzido quimicamente , Substância Negra/fisiopatologia , Sinapses/efeitos dos fármacos , Trombina/toxicidade , Animais , Apoptose/fisiologia , Corpo Estriado/patologia , Corpo Estriado/fisiopatologia , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Feminino , Glutamato Descarboxilase/metabolismo , Glicoproteínas de Membrana/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/efeitos dos fármacos , Vias Neurais/patologia , Vias Neurais/fisiopatologia , Neurotoxinas/toxicidade , Doença de Parkinson/patologia , Doença de Parkinson/fisiopatologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/patologia , Proteínas Proto-Oncogênicas c-fos/genética , Ratos , Ratos Wistar , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/metabolismo , Degeneração Retrógrada/patologia , Degeneração Retrógrada/fisiopatologia , Estilbamidinas , Substância Negra/metabolismo , Substância Negra/patologia , Sinapses/metabolismo , Sinapses/patologia , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/metabolismo , Sinaptofisina/efeitos dos fármacos , Sinaptofisina/metabolismo , Tirosina 3-Mono-Oxigenase/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/metabolismo
19.
Neurogastroenterol Motil ; 20(4): 385-91, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18179608

RESUMO

Proteinase-activated receptor-1 (PAR(1)) and PAR(2) mediate contraction in the guinea-pig gallbladder. To investigate and compare the effects mediated by PARs in the human gallbladder with those in the guinea-pig gallbladder, we measured contractions of isolated human and guinea-pig gallbladder strips caused by PAR agonists. Results in human were similar to those in guinea-pig gallbladder. The PAR(1) agonists, thrombin, TFLLR-NH2 and SFLLRN-NH2, as well as the PAR(2) agonists, trypsin, SLIGKV-NH2 and SLIGRL-NH2, caused contraction in both human and guinea-pig gallbladders. These indicate the existence of PAR(1) and PAR(2) mediating gallbladder contraction. Furthermore, the existence of PAR(1) and PAR(2) in the human gallbladder was confirmed by reverse transcription-polymerase chain reaction. In contrast, FSLLR-NH2, a PAR(1) control peptide, and VKGILS-NH2, a PAR(2) control peptide, as well as three PAR(4) agonists, GYPGKF-NH2, GYPGQV-NH2 and AYPGKF-NH2, did not cause any contraction or relaxation. The contractile responses to TFLLR-NH2, SFLLRN-NH2 and trypsin in both human and guinea-pig gallbladders were insensitive to atropine and tetrodotoxin, suggesting direct effects. These results demonstrate that, similar to the guinea-pig gallbladder, both PAR(1) and PAR(2) but not PAR(4) mediate muscle contraction in the human gallbladder. PAR(1) and PAR(2) may play important roles in the control of both human and guinea-pig gallbladder motility.


Assuntos
Vesícula Biliar/metabolismo , Contração Muscular/fisiologia , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Receptores de Trombina/metabolismo , Animais , Vesícula Biliar/efeitos dos fármacos , Esvaziamento da Vesícula Biliar/fisiologia , Cobaias , Humanos , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Liso/fisiologia , Técnicas de Cultura de Órgãos , RNA Mensageiro/análise , Receptor PAR-1/efeitos dos fármacos , Receptor PAR-2/efeitos dos fármacos , Receptores de Trombina/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trombina/farmacologia , Tripsina/farmacologia
20.
Glia ; 55(9): 917-25, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17437307

RESUMO

High concentrations of thrombin (Thr) have been linked to neuronal damage in cerebral ischemia and traumatic brain injury. In the present study we found that Thr markedly enhanced swelling-activated efflux of (3)H-glutamate from cultured astrocytes exposed to hyposmotic medium. Thr (0.5-5 U/mL) elicited small (3)H-glutamate efflux under isosmotic conditions and increased the hyposmotic glutamate efflux by 5- to 10-fold, the maximum effect being observed at 15% osmolarity reduction. These Thr effects involve its protease activity and are fully mimicked by SFFLRN, the synthetic peptide activating protease-activated receptor-1. Thr potentiation of (3)H-glutamate efflux was largely dependent on a Thr-elicited increases in cytosolic Ca(2+) (Ca(2+) (i)) concentration ([Ca(2+)](i)). Preventing Ca(2+) (i) rise by treatment with EGTA-AM or with the phospholipase C blocker U73122 reduced the Thr-increased glutamate efflux by 68%. The protein kinase C blockers Go6976 or chelerythrine reduced the Thr effect by 19%-22%, while Ca/calmodulin blocker W7 caused a 63% inhibition. In addition to this Ca(2+)-sensitive pathway, Thr effect on glutamate efflux also involved activation of phosphoinositide-3 kinase (PI3K), since it was reduced by the PI3K inhibitor wortmannin (51% inhibition). Treating cells with EGTA-AM plus wortmannin essentially abolished Thr-dependent glutamate efflux. Thr-activated glutamate release was potently inhibited by the blockers of the volume-sensitive anion permeability pathway, NPPB (IC(50) 15.8 microM), DCPIB (IC(50) 4.2 microM), and tamoxifen (IC(50) 6.6 microM. These results suggest that Thr may contribute to the excitotoxic neuronal injury by elevating extracellular glutamate release from glial cells. Therefore, this work may aid in search of neuroprotective strategies for treating cerebral ischemia and brain trauma.


Assuntos
Astrócitos/metabolismo , Dano Encefálico Crônico/metabolismo , Edema Encefálico/metabolismo , Ácido Glutâmico/metabolismo , Trombina/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Dano Encefálico Crônico/fisiopatologia , Edema Encefálico/fisiopatologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Líquido Extracelular/metabolismo , Soluções Hipotônicas/farmacologia , Oligopeptídeos/farmacologia , Pressão Osmótica/efeitos dos fármacos , Ratos , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Trombina/farmacologia , Equilíbrio Hidroeletrolítico/efeitos dos fármacos , Equilíbrio Hidroeletrolítico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...