Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 105(25): 8754-9, 2008 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-18559859

RESUMO

The amyloid hypothesis states that a variety of neurotoxic beta-amyloid (Abeta) species contribute to the pathogenesis of Alzheimer's disease. Accordingly, a key determinant of disease onset and progression is the appropriate balance between Abeta production and clearance. Enzymes responsible for the degradation of Abeta are not well understood, and, thus far, it has not been possible to enhance Abeta catabolism by pharmacological manipulation. We provide evidence that Abeta catabolism is increased after inhibition of plasminogen activator inhibitor-1 (PAI-1) and may constitute a viable therapeutic approach for lowering brain Abeta levels. PAI-1 inhibits the activity of tissue plasminogen activator (tPA), an enzyme that cleaves plasminogen to generate plasmin, a protease that degrades Abeta oligomers and monomers. Because tPA, plasminogen and PAI-1 are expressed in the brain, we tested the hypothesis that inhibitors of PAI-1 will enhance the proteolytic clearance of brain Abeta. Our data demonstrate that PAI-1 inhibitors augment the activity of tPA and plasmin in hippocampus, significantly lower plasma and brain Abeta levels, restore long-term potentiation deficits in hippocampal slices from transgenic Abeta-producing mice, and reverse cognitive deficits in these mice.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Fibrinolisina/metabolismo , Fibrinolíticos/metabolismo , Animais , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Inativadores de Plasminogênio/metabolismo , Ativador de Plasminogênio Tecidual/antagonistas & inibidores , Ativador de Plasminogênio Tecidual/metabolismo
2.
Thromb Haemost ; 99(4): 749-58, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18392333

RESUMO

This study aimed to evaluate a small-molecule PAI-1 inhibitor (PAI-039; tiplaxtinin) in a rodent stenosis model of venous thrombosis in a two-phase experiment. Phase 1 determined the efficacy of tiplaxtinin against Lovenox (LOV), while phase 2 determined the dose-dependent efficacy. For both phases, drug treatment began 24 hours after surgically induced venous thrombosis and continued for four days. Phase 1 animals (n = 24) receiving low-dose (LD; 1 mg/kg oral gavage) PAI-1 inhibitor demonstrated a 52% decrease in thrombus weight (TW) versus controls (p < 0.05) with significant reductions in active plasma PAI-1, while the high-dose (HD; 10 mg/kg oral gavage) group demonstrated a 23% reduction in TW versus controls. Animals treated subcutaneously with LOV (3 mg/kg) showed a 39% decrease in TW versus controls (p < 0.05). Coagulation tests (aPTT and TCT) were significantly different in LOV compared to PAI-1 inhibitor groups. PAI-039 treatment was also associated with significantly increased return of inferior vena cava blood flow four days post-thrombosis versus controls (p < 0.05). In phase 2 (n = 30), TW was reduced from the 0.5 mg/kg to 5 mg/kg experimental groups, with the 10 mg/kg group demonstrating a paradoxical increase. The 5 mg/kg group showed statistically significant decreases in TW versus controls after four treatment days (p < 0.05). This is the first study to demonstrate dose related effects of PAI-039 on increasing thrombus resolution and inferior vena cava blood flow without adverse effects on anti-coagulation in a rat stenosis model of venous thrombosis.


Assuntos
Ácidos Indolacéticos/administração & dosagem , Inibidor 1 de Ativador de Plasminogênio/sangue , Trombose Venosa/sangue , Trombose Venosa/tratamento farmacológico , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Enoxaparina/antagonistas & inibidores , Fibrose , Inflamação/tratamento farmacológico , Inflamação/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Túnica Íntima/patologia , Veia Cava Inferior/patologia , Trombose Venosa/patologia
3.
Arterioscler Thromb Vasc Biol ; 25(2): 365-71, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15576638

RESUMO

OBJECTIVE: To test the hypothesis that pharmacological plasminogen activator inhibitor (PAI)-1 inhibition protects against renin-angiotensin-aldosterone system-induced cardiovascular injury, the effect of a novel orally active small-molecule PAI-1 inhibitor, PAI-039, was examined in a mouse model of angiotensin (Ang) II-induced vascular remodeling and cardiac fibrosis. METHODS AND RESULTS: Uninephrectomized male C57BL/6J mice were randomized to vehicle subcutaneus, Ang II (1 mug/h) subcutaneous, vehicle+PAI-039 (1 mg/g chow), or Ang II+PAI-039 during high-salt intake for 8 weeks. Ang II caused significant medial, adventitial, and aortic wall thickening compared with vehicle. PAI-039 attenuated Ang II-induced aortic remodeling without altering the pressor response to Ang II. Ang II increased heart/body weight ratio and cardiac fibrosis. PAI-039 did not attenuate the effect of Ang II on cardiac hypertrophy and increased fibrosis. The effect of PAI-039 on Ang II/salt-induced aortic remodeling and cardiac fibrosis was comparable to the effect of genetic PAI-1 deficiency. Ang II increased aortic mRNA expression of PAI-1, collagen I, collagen III, fibronectin, osteopontin, monocyte chemoattractant protein-1, and F4/80; PAI-039 significantly decreased the Ang II-induced increase in aortic osteopontin expression at 8 weeks. CONCLUSIONS: This study demonstrates that pharmacological inhibition of PAI-1 protects against Ang II-induced aortic remodeling. Future studies are needed to determine whether the interactive effect of Ang II/salt and reduced PAI-1 activity on cardiac fibrosis is species-specific. In this study, the effect of pharmacological PAI-1 inhibition in a mouse model of Ang II-induced vascular remodeling and cardiac fibrosis was examined. PAI-1 inhibition significantly attenuated Ang II-induced aortic medial and wall thickening, but not cardiac hypertrophy, and enhanced Ang II/salt-induced cardiac fibrosis.


Assuntos
Acetatos/uso terapêutico , Angiotensina II/toxicidade , Aorta/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Indóis/uso terapêutico , Rim/efeitos dos fármacos , Miocárdio/patologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Cloreto de Sódio na Dieta/toxicidade , Acetatos/farmacologia , Administração Oral , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/induzido quimicamente , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Pressão Sanguínea/efeitos dos fármacos , Quimiocina CCL2/biossíntese , Quimiocina CCL2/genética , Colágeno Tipo I/biossíntese , Colágeno Tipo I/genética , Colágeno Tipo III/biossíntese , Colágeno Tipo III/genética , Avaliação Pré-Clínica de Medicamentos , Fibronectinas/biossíntese , Fibronectinas/genética , Fibrose , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Glomerulosclerose Segmentar e Focal/patologia , Glomerulosclerose Segmentar e Focal/prevenção & controle , Hipertrofia Ventricular Esquerda/induzido quimicamente , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/prevenção & controle , Ácidos Indolacéticos , Indóis/farmacologia , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Nefrectomia , Osteopontina , Inibidor 1 de Ativador de Plasminogênio/deficiência , Inibidor 1 de Ativador de Plasminogênio/genética , RNA Mensageiro/biossíntese , Distribuição Aleatória , Sialoglicoproteínas/biossíntese , Sialoglicoproteínas/genética , Método Simples-Cego
4.
J Med Chem ; 47(14): 3491-4, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15214776

RESUMO

Indole oxoacetic acid derivatives were prepared and evaluated for in vitro binding to and inactivation of human plasminogen activator inhibitor-1 (PAI-1). SAR based on biochemical, physiological, and pharmacokinetic attributes led to identification of tiplaxtinin as the optimal selective PAI-1 inhibitor. Tiplaxtinin exhibited in vivo oral efficacy in two different models of acute arterial thrombosis. The remarkable preclinical safety and metabolic stability profiles of tiplaxtinin led to advancing the compound to clinical trials.


Assuntos
Indóis/síntese química , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidores de Serina Proteinase/síntese química , Administração Oral , Animais , Disponibilidade Biológica , Trombose das Artérias Carótidas/tratamento farmacológico , Trombose Coronária/tratamento farmacológico , Cães , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Ácidos Indolacéticos , Indóis/química , Indóis/farmacologia , Ratos , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/farmacologia , Relação Estrutura-Atividade
5.
J Med Chem ; 47(3): 681-95, 2004 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-14736248

RESUMO

A novel series of substituted sulfanyldihydroimidazolones (1) that modulates high-density lipoprotein cholesterol (HDL-C) has been reported to have HDL-elevating properties in several animal models. Concerns about the chemical and metabolic stability of 1 directed us to explore the structure-activity relationship (SAR) of a related series of substituted thiohydantoins (2). Expansion of the scope of the thiohydantoin series led to exploration of compounds in related thio-containing ring systems 3-7 and the N-cyanoguanidine derivative 8. Compounds were tested sequentially in three animal models to assess their HDL-C elevating efficacy and safety profiles. Further evaluation of selected compounds in a dose-response paradigm culminated in the identification of compound 2.39 as a candidate compound for advanced preclinical studies.


Assuntos
HDL-Colesterol/sangue , Imidazóis/síntese química , Tioidantoínas/síntese química , Tionas/síntese química , Administração Oral , Animais , Cricetinae , Desenho de Fármacos , Guanidinas/síntese química , Guanidinas/química , Guanidinas/farmacologia , Hipercolesterolemia/sangue , Imidazóis/química , Imidazóis/farmacologia , Masculino , Piperazinas/síntese química , Piperazinas/química , Piperazinas/farmacologia , Pirimidinas/síntese química , Pirimidinas/química , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Estimulação Química , Relação Estrutura-Atividade , Tiazolidinedionas/síntese química , Tiazolidinedionas/química , Tiazolidinedionas/farmacologia , Tioidantoínas/química , Tioidantoínas/farmacologia , Tionas/química , Tionas/farmacologia
6.
Mol Pharmacol ; 72(4): 897-906, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17622579

RESUMO

PAI-749 is a potent and selective synthetic antagonist of plasminogen activator inhibitor 1 (PAI-1) that preserved tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) activities in the presence of PAI-1 (IC(50) values, 157 and 87 nM, respectively). The fluorescence (Fl) of fluorophore-tagged PAI-1 (PAI-NBD119) was quenched by PAI-749; the apparent K(d) (254 nM) was similar to the IC(50) (140 nM) for PAI-NBD119 inactivation. PAI-749 analogs displayed the same potency rank order for neutralizing PAI-1 activity and perturbing PAI-NBD119 Fl; hence, binding of PAI-749 to PAI-1 and inactivation of PAI-1 activity are tightly linked. Exposure of PAI-1 to PAI-749 for 5 min (sufficient for full inactivation) followed by PAI-749 sequestration with Tween 80 micelles yielded active PAI-1; thus, PAI-749 did not irreversibly inactivate PAI-1, a known metastable protein. Treatment of PAI-1 with a PAI-749 homolog (producing less assay interference) blocked the ability of PAI-1 to displace p-aminobenzamidine from the uPA active site. Consistent with this observation, PAI-749 abolished formation of the SDS-stable tPA/PAI-1 complex. PAI-749-mediated neutralization of PAI-1 was associated with induction of PAI-1 polymerization as assessed by native gel electrophoresis. PAI-749 did not turn PAI-1 into a substrate for tPA; however, PAI-749 promoted plasmin-mediated degradation of PAI-1. In conclusion, PAI-1 inactivation by PAI-749 using purified components can result from a dual mechanism of action. First, PAI-749 binds directly to PAI-1, blocks PAI-1 from accessing the active site of tPA, and abrogates formation of the SDS-stable tPA/PAI-1 complex. Second, binding of PAI-749 to PAI-1 renders PAI-1 vulnerable to plasmin-mediated proteolytic degradation.


Assuntos
Indóis/farmacologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Tetrazóis/farmacologia , Biopolímeros/metabolismo , Eletroforese em Gel de Poliacrilamida , Fluorescência , Humanos , Hidrólise , Micelas , Vitronectina/metabolismo , Vitronectina/farmacologia
7.
J Biol Chem ; 282(12): 9288-96, 2007 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-17276980

RESUMO

The inactivation of plasminogen activator inhibitor-1 (PAI-1) by the small molecule PAI-1 inhibitor PAI-039 (tiplaxtinin) has been investigated using enzymatic analysis, direct binding studies, site-directed mutagenesis, and molecular modeling studies. Previously PAI-039 has been shown to exhibit in vivo activity in various animal models, but the mechanism of inhibition is unknown. PAI-039 bound specifically to the active conformation of PAI-1 and exhibited reversible inactivation of PAI-1 in vitro. SDS-PAGE indicated that PAI-039 inactivated PAI-1 predominantly through induction of PAI-1 substrate behavior. Preincubation of PAI-1 with vitronectin, but not bovine serum albumin, blocked PAI-039 activity while analysis of the reciprocal experiment demonstrated that preincubation of PAI-1 with PAI-039 blocked the binding of PAI-1 to vitronectin. Together, these data suggest that the site of interaction of the drug on PAI-1 is inaccessible when PAI-1 is bound to vitronectin and may overlap with the PAI-1 vitronectin binding domain. This was confirmed by site-directed mutagenesis and molecular modeling studies, which suggest that the binding epitope for PAI-039 is localized adjacent to the previously identified interaction site for vitronectin. Thus, these studies provide a detailed characterization of the mechanism of inhibition of PAI-1 by PAI-039 against free, but not vitronectin-bound PAI-1, suggesting for the first time a novel pool of PAI-1 exists that is vulnerable to inhibition by inactivators that bind at the vitronectin binding site.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/química , Sítios de Ligação , Relação Dose-Resposta a Droga , Glicosilação , Humanos , Ácidos Indolacéticos/farmacologia , Concentração Inibidora 50 , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície , Fatores de Tempo , Vitronectina/química
8.
Bioorg Med Chem ; 15(18): 6208-26, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17624788

RESUMO

1-Aminoethyl-3-arylsulfonyl-1H-indoles 1 are 5-HT(6) receptor ligands with modest activity in a 5-HT(6) cyclase assay. Introduction of an additional nitrogen in the indole ring provides 1-aminoethyl-3-arylsulfonyl-1H-pyrrolo[2,3-b]pyridines 2 with both enhanced 5-HT(6) affinity and cyclase activity, many acting as 5-HT(6) agonists. We constrained the basic side chain as part of a ring to make 1-(azacyclyl)-3-arylsulfonyl-1H-pyrrolo[2,3-b]pyridines incorporating a pyrrolidinyl 3 or piperidinyl 4 ring system. Preparation of compounds 3 and 4 required synthesis of the key intermediates, 1-(pyrrolidin-3-yl)-1H-pyrrolo[2,3-b]pyridines 7 and 1-(piperidin-3-yl)-1H-pyrrolo[2,3-b]pyridines 8, respectively. Intermediates 7 were prepared through alkylation of 7-azaindole while the intermediates 8 required an alternate synthesis. The compounds of both series 3 and 4 were shown to have high binding affinities for the 5-HT(6) receptor. The in vitro functional activity at the 5-HT(6) receptor varied depending on various functionalities including the selection of the arylsulfonyl, the substitution on the arylsulfonyl group, the ring size, and the substitution on the basic amine moiety producing either 5-HT(6) receptor agonists or antagonists.


Assuntos
Piridinas/farmacologia , Receptores de Serotonina/química , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Ligação Competitiva , Células HeLa/efeitos dos fármacos , Humanos , Ligantes , Estrutura Molecular , Piridinas/síntese química , Piridinas/química , Receptores de Serotonina/metabolismo , Antagonistas da Serotonina/síntese química , Antagonistas da Serotonina/química , Agonistas do Receptor de Serotonina/síntese química , Agonistas do Receptor de Serotonina/química , Relação Estrutura-Atividade
9.
Blood ; 107(1): 132-4, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16160004

RESUMO

Hepatic veno-occlusive disease (VOD) is a common complication of high-dose chemotherapy associated with bone marrow transplantation. While the pathogenesis of VOD is uncertain, plasminogen activator inhibitor-1 (PAI-1) has emerged as a diagnostic marker and predictor of VOD in humans. In this study, we investigated the role of PAI-1 in a murine model of VOD produced by long-term nitric oxide synthase inhibition using L-NAME. After 6 weeks, wild-type (WT) mice developed extensive fibrinoid hepatic venous thrombi and biochemical evidence of hepatic injury and dysfunction. In contrast, PAI-1-deficient mice were largely protected from the development of hepatic vein thrombosis. Furthermore, WT mice that received tiplaxtinin, an antagonist of PAI-1, were effectively protected from L-NAME-induced thrombosis. Taken together, these data indicate that NO and PAI-1 play pivotal and antagonistic roles in hepatic vein thrombosis and that PAI-1 is a potential target in the prevention and treatment of VOD in humans.


Assuntos
Síndrome de Budd-Chiari/etiologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Animais , Síndrome de Budd-Chiari/induzido quimicamente , Síndrome de Budd-Chiari/prevenção & controle , Modelos Animais de Doenças , Hepatopatia Veno-Oclusiva/induzido quimicamente , Hepatopatia Veno-Oclusiva/etiologia , Ácidos Indolacéticos , Indóis/farmacologia , Camundongos , Camundongos Knockout , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Inibidor 1 de Ativador de Plasminogênio/genética
10.
J Pharmacol Exp Ther ; 314(2): 710-6, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15860572

RESUMO

We tested a novel, orally active inhibitor of plasminogen activator inhibitor-1 (PAI-1) in a canine model of electrolytic injury. Dogs received by oral gavage either vehicle (control) or the PAI-1 inhibitor PAI-039 [{1-benzyl-5-[4-(trifluoromethoxy)phenyl]-1H-indol-3-yl}(oxo)acetic acid] (1, 3, and 10 mg/kg) and were subjected to electrolytic injury of the coronary artery. PAI-039 caused prolongation in time to coronary occlusion (control, 31.7 +/- 6.3 min; 3 mg/kg PAI-039, 66.0 +/- 6.4 min; 10 mg/kg, 56.7 +/- 7.4 min; n = 5-6; p < 0.05) and a reduced thrombus weight (control, 7.6 +/- 1.5 mg; 10 mg/kg PAI-039, 3.6 +/- 1.0 mg; p < 0.05). Although occlusive thrombosis was observed across all groups based upon the absence of measurable blood flow, a high incidence (>60%) of spontaneous reperfusion occurred only in those groups receiving PAI-039. Spontaneous reperfusion in the 10 mg/kg PAI-039 group accounted for total blood flow (area under the curve of coronary blood flow) of 99.6 +/- 11.7 ml after initial thrombotic occlusion (p < 0.05 compared with control). Plasma PAI-1 activity was reduced in all drug-treated groups (percentage of reduction in activity p < 0.05; 10 mg/kg PAI-039), whereas ADP-, 9,11-dideoxy-11alpha,9alpha-epoxymethanoprostaglandin F(2alpha) (U46619)-, and collagen-induced platelet aggregation, as well as template bleeding and prothrombin time, remained unaffected by PAI-039. Ex vivo clot lysis analysis revealed normal clot formation but accelerated clot lysis in PAI-039-treated groups. The pharmacokinetic profile of PAI-039 indicated an oral bioavailability of 43 +/- 15.3% and a plasma half-life of 6.2 +/- 1.3 h. In conclusion, PAI-039 is an orally active prothrombolytic drug that inhibits PAI-1 and accelerates fibrinolysis while maintaining normal coagulation in a model of coronary occlusion.


Assuntos
Acetatos/uso terapêutico , Trombose Coronária/tratamento farmacológico , Fibrinolíticos/uso terapêutico , Indóis/uso terapêutico , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Acetatos/sangue , Acetatos/farmacologia , Animais , Tempo de Sangramento , Pressão Sanguínea/efeitos dos fármacos , Circulação Coronária/efeitos dos fármacos , Cães , Determinação de Ponto Final , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/sangue , Fibrinolíticos/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Hemostasia/efeitos dos fármacos , Ácidos Indolacéticos , Indóis/sangue , Indóis/farmacologia , Tempo de Tromboplastina Parcial , Agregação Plaquetária/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
11.
J Biol Chem ; 278(18): 16329-35, 2003 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-12606560

RESUMO

The mechanism for the conversion of plasminogen activator inhibitor-1 (PAI-1) from the active to the latent conformation is not well understood. Recently, a monoclonal antibody, 33B8, was described that rapidly converts PAI-1 to the latent conformation (Verhamme, I., Kvassman, J. O., Day, D., Debrock, S., Vleugels, N., Declerck, P. J., and Shore, J. D. (1999) J. Biol. Chem. 274, 17511-17517). In an attempt to understand this interaction, and more broadly to understand the mechanism of the natural transition of PAI-1 to the latent conformation, we have used random mutagenesis to identify the 33B8 epitope in PAI-1. This site involves at least 8 amino acids scattered over more than two-thirds of the linear sequence that form a compact epitope on the PAI-1 three-dimensional structure. Surface plasmon resonance studies indicate a high affinity interaction between latent PAI-1 and 33B8 that is approximately 100-fold higher than comparable binding to active PAI-1. Structural modeling results together with surface plasmon resonance analysis of parental and site-directed PAI-1 mutants with disrupted 33B8 binding suggest the existence of a specific PAI-1 intermediate structure that is stabilized by 33B8 binding. These analyses strongly suggest that this intermediate form of PAI-1 has a partial insertion of the reactive center loop into beta-sheet A, and together, these data have significant implications for the general serpin mechanism of proteinase inhibition.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/química , Serpinas/fisiologia , Animais , Anticorpos Monoclonais/imunologia , Mapeamento de Epitopos , Camundongos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Inibidor 1 de Ativador de Plasminogênio/imunologia , Conformação Proteica , Análise de Sequência de DNA , Ressonância de Plasmônio de Superfície
12.
Bioorg Med Chem Lett ; 12(15): 1967-71, 2002 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12113820

RESUMO

Three novel metabolites of the angiotensin-II (A-II) receptor antagonist tasosartan have been identified in humans, and the syntheses and pharmacologic profiling of these metabolites are reported. Each metabolite bound the human A-II receptor with IC(50)s between 20 and 45nM. The in vivo effects of these compounds in attenuating the pressor response to angiotensin-II challenge in anesthetized rats were also investigated. An unsaturated diol metabolite exhibited in vivo efficacy at intravenous doses of 1 and 3mg/kg, while the other metabolites, both carboxylic acids, had no significant effect at the same doses.


Assuntos
Antagonistas de Receptores de Angiotensina , Pirimidinas/metabolismo , Pirimidinas/farmacologia , Tetrazóis/metabolismo , Tetrazóis/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Frequência Cardíaca/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Injeções Intravenosas , Pirimidinas/química , Ratos , Receptores de Angiotensina/metabolismo , Relação Estrutura-Atividade , Tetrazóis/química
13.
Biochem Biophys Res Commun ; 311(4): 904-8, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-14623266

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is the major physiological inhibitor of tissue plasminogen activator (tPA) and is elevated in diseases of vascular remodeling. In this study, we describe an inhibitor of active PAI-1, WAY-140312. Using fluorescence spectroscopy, it was determined that WAY-140312 bound PAI-1 at a single binding site with a dissociation constant of 5 microM. In a biochemical assay determining direct tPA activity, human recombinant PAI-1 completely inhibited tPA, but this inhibition was blocked by WAY-140312 at an IC(50) of 15.6 microM. In vivo, a 10 mg/kg oral dose of WAY-140312 to rats produced a significant plasma reduction of active PAI-1. Bleeding time, thrombin clotting time, and ex vivo platelet aggregation induced by ADP (20 microM) or collagen (2.5 microg/ml) were not affected by administration of WAY-140312. These results are the first to demonstrate that an orally active PAI-1 inhibitor can reduce plasma PAI-1 activity while maintaining normal platelet aggregation and coagulation.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Hemorragia/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Administração Oral , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Hemorragia/sangue , Humanos , Masculino , Inibidor 1 de Ativador de Plasminogênio/sangue , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa