Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Carbohydr Polym ; 222: 115025, 2019 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-31320079

RESUMO

Fucosylated chondroitin sulfate (FCS) oligosaccharides of specific molecular weight have shown potent anticoagulant activities with selectivity towards intrinsic factor Xase complex. However, the preparation of FCS oligosaccharides by traditional methods requires multiple purification steps consuming large amounts of time and significant resources. The current study focuses on developing a method for the rapid preparation of FCS oligomers from sea cucumber Pearsonothuria graeffei having 6-18 saccharide residues. The key steps controlling molecular weight (Mw) and purity of these FCS oligomers were evaluated. Structural analysis showed the resulting FCS oligomers were primarily l-Fuc3,4diS-α1,3-d-GlcA-ß1,3-(d-GalNAc4,6diS-ß1,4-[l-Fuc3,4diS-α1,3-]d-GlcA-ß1,3-)nd-anTal-ol4,6diS (n = 1˜5) accompanied by partial de-fucosylation and/or de-sulfation. In vitro and in vivo experiments demonstrate that these FCS oligomers selectively inhibit intrinsic factor Xase complex and exhibit remarkable antithrombotic activity without hemorrhagic and hypotension side effects. This method is suitable for large-scale preparation of FCS oligosaccharides as clinical anticoagulants.


Assuntos
Anticoagulantes/uso terapêutico , Sulfatos de Condroitina/uso terapêutico , Fator IXa/antagonistas & inibidores , Fator VIIIa/antagonistas & inibidores , Fibrinolíticos/uso terapêutico , Proteínas de Neoplasias/antagonistas & inibidores , Animais , Anticoagulantes/química , Anticoagulantes/farmacologia , Sequência de Carboidratos , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Cisteína Endopeptidases , Fibrinolíticos/química , Fibrinolíticos/farmacologia , Masculino , Camundongos , Coelhos , Ratos Sprague-Dawley , Pepinos-do-Mar/química , Trombose Venosa/tratamento farmacológico
2.
Carbohydr Res ; 467: 45-51, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30114596

RESUMO

Fucosylated chondroitin sulfate (FuCS) is a structurally complex glycosaminoglycan found in sea cucumbers with a wide spectrum of biological activities, among which anticoagulant activity is particularly attractive for the development of alternative anticoagulant drugs with decreased adverse effects and risks of bleeding. Previous studies show that FuCS glycomimetics bearing several trisaccharide epitopes displayed promising anticoagulant activity and did not change the mode of action of FuCS. To simplify synthetic difficulty of high valent glycoclusters and obtain candidate compounds with relatively low molecular weights, here we report the synthesis of two FuCS glycoclusters with low valence and more compact structures. Anticoagulation studies showed that these simplified "short-armed" glycoclusters demonstrated comparable potency with "long-armed" high valent glycoclusters, offering a concise approach for the development of novel anticoagulant agents.


Assuntos
Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Sulfatos de Condroitina/farmacologia , Inibidores Enzimáticos/farmacologia , Glicosídeos/farmacologia , Anticoagulantes/síntese química , Anticoagulantes/química , Configuração de Carboidratos , Sulfatos de Condroitina/síntese química , Sulfatos de Condroitina/química , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Fator IXa/antagonistas & inibidores , Fator IXa/metabolismo , Fator VIIIa/antagonistas & inibidores , Fator VIIIa/metabolismo , Glicosídeos/síntese química , Glicosídeos/química , Humanos
3.
Arterioscler Thromb Vasc Biol ; 32(10): 2387-93, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22904276

RESUMO

OBJECTIVE: Protein S is a vitamin K-dependent plasma protein that functions in the feedback regulation of thrombin generation. Our goal was to determine how protein S regulates the intrinsic pathway of blood coagulation. METHODS AND RESULTS: We used plasma, including platelet-rich plasma, and in vitro methods to determine how the intrinsic pathway of blood coagulation is regulated by protein S. We obtained the following results: (1) activated partial thromboplastin time assays with protein S-supplemented plasma confirmed that protein S prolongs clotting time; (2) a modified activated partial thromboplastin time assay with factor IX (fIX)-deficient plasma confirmed that protein S affects fIX-initiated clotting; (3) a fIXa/factor VIIIa (fVIIIa)-mediated thrombin generation assay with either platelet-rich plasma or factor-deficient plasma, initiated with a limiting amount of tissue factor, was regulated by protein S; (4) in the presence of phosphatidylserine vesicles, protein S inhibited fIXa in the absence and presence of fVIIIa; and (5) protein S altered only the K(M) for factor X activation by fIXa in the absence of fVIIIa and both k(cat) and K(M) in the presence of fVIIIa. CONCLUSIONS: From our findings, it can be concluded that protein S inhibits fIXa in the presence or absence of fVIIIa in an activated protein C-independent way.


Assuntos
Coagulação Sanguínea/fisiologia , Fator IXa/antagonistas & inibidores , Fator VIIIa/antagonistas & inibidores , Proteína C/fisiologia , Proteína S/fisiologia , Fator IXa/fisiologia , Fator VIIIa/fisiologia , Retroalimentação Fisiológica/fisiologia , Humanos , Técnicas In Vitro , Tempo de Tromboplastina Parcial , Transdução de Sinais/fisiologia , Trombina/fisiologia
4.
Thromb Haemost ; 105(6): 968-80, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21475777

RESUMO

Mechanisms of inflammation and coagulation are linked through various pathways. Human neutrophil elastase (HNE), can bind to activated platelets, might be localised on platelet membranes that provide negatively-charged phospholipid essential for the optimum function of tenase complex. In this study, we examined the effect of HNE on factor (F)VIII. FVIII activity was rapidly diminished in the presence of HNE and was undetectable within 10 minutes. The inactivation rate was ~8-fold greater than that of activated protein C (APC). This time-dependent inactivation was moderately affected by von Willebrand factor. HNE proteolysed the heavy chain (HCh) of FVIII into two terminal products, A11-358 and A2375-708, by limited proteolysis at Val358, Val374, and Val708. Cleavage at Val708 was much slower than that at Val358 in the >90-kDa A1-A2-B compared to the 90-kDa A1-A2. The 80-kDa light chain (LCh) was proteolysed to 75-kDa product by cleavage at Val1670. HNE-catalysed FVIIIa inactivation was markedly slower than that of native FVIII (by ~25-fold), due to delayed cleavage at Val708 in FVIIIa. The inactivation rate mediated by HNE was ~8-fold lower than that by APC. Cleavages at Val358 and Val708 were regulated by the presence of LCh and HCh, respectively. In conclusion, HNE-catalysed FVIII inactivation was associated with the limited-proteolysis that led to A11-358, A2375-708, and A3-C1-C21671-2332, and subsequently to critical cleavage at Val708. HNE-related FVIII(a) reaction might play a role in inactivation of HNE-induced coagulation process, and appeared to depend on the amounts of inactivated FVIII and active FVIIIa which is predominantly resistant to HNE inactivation.


Assuntos
Fator VIIIa/antagonistas & inibidores , Elastase de Leucócito/farmacologia , Peptídeo Hidrolases/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Catálise/efeitos dos fármacos , Fator VIIIa/metabolismo , Fibrinolisina/metabolismo , Humanos , Hidrólise/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Fator de von Willebrand/metabolismo
6.
Blood ; 110(13): 4234-42, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17848617

RESUMO

The diversity of factor VIII (fVIII) C2 domain antibody epitopes was investigated by competition enzyme-linked immunosorbent assay (ELISA) using a panel of 56 antibodies. The overlap patterns produced 5 groups of monoclonal antibodies (MAbs), designated A, AB, B, BC, and C, and yielded a set of 18 distinct epitopes. Group-specific loss of antigenicity was associated with mutations at the Met2199/Phe2200 phospholipid binding beta-hairpin (group AB MAbs) and at Lys2227 (group BC MAbs), which allowed orientation of the epitope structure as a continuum that covers one face of the C2 beta-sandwich. MAbs from groups A, AB, and B inhibit the binding of fVIIIa to phospholipid membranes. Group BC was the most common group and displayed the highest specific fVIII inhibitor activities. MAbs in this group are type II inhibitors that inhibit the activation of fVIII by either thrombin or factor Xa and poorly inhibit the binding of fVIII to phospholipid membranes or von Willebrand factor (VWF). Group BC MAbs are epitopically and mechanistically distinct from the extensively studied group C MAb, ESH8. These results reveal the structural and functional complexity of the anti-C2 domain antibody response and indicate that interference with fVIII activation is a major attribute of the inhibitor landscape.


Assuntos
Anticorpos Monoclonais/imunologia , Mapeamento de Epitopos/métodos , Epitopos/imunologia , Fator VIII/imunologia , Fator VIIIa/antagonistas & inibidores , Hemofilia A/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Fator VIII/antagonistas & inibidores , Fator VIII/química , Humanos , Camundongos , Fosfolipídeos/metabolismo , Fator de von Willebrand/metabolismo
7.
Acta Haematol ; 115(3-4): 186-91, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16549894

RESUMO

Alpha1-antitrypsin (alpha1-AT) is a physiological inhibitor of activated protein C (APC) and therefore decreased APC activity. APC itself causes an anticoagulant effect by inactivating factors Va and VIIIa. The present case-control study was performed to evaluate the role of the elevated alpha1-AT concentration in pediatric patients with ischemic stroke (IS). alpha1-AT concentrations were measured along with established prothrombotic risk factors 6-12 months after the acute thrombotic onset in 81 Caucasian children with IS aged 1 month to 18 years. The cutoff values defined as age-dependent 90th percentiles were obtained from 229 healthy controls. Median (range) values of alpha1-AT were significantly higher in patients compared with control subjects [122.0 mg/dl (61.4-224.0) vs. 114.0 mg/dl (66.8-156.0); p = 0.016]. In addition, 14 of the 81 patients (17.3%) compared with 10 of the 162 controls (6.2%) had alpha1-AT concentrations above the 90th age-dependent percentiles (p = 0.012). Multivariate analysis performed in a 1:2 matched case-control setting adjusted for the presence of established prothrombotic risk factors showed a significantly increased odds ratio (OR) and 95% confidence interval (CI) for patients with elevated alpha1-AT >90th percentiles and IS (OR/CI: 4.0/1.64-9.92; p = 0.0024). Data shown here give evidence that total alpha1-AT concentrations above the 90th age-dependent percentiles independently increase the risk of IS 4.0-fold in Caucasian children.


Assuntos
Infarto Cerebral/sangue , alfa 1-Antitripsina/análise , Adolescente , Fatores Etários , Estudos de Casos e Controles , Criança , Pré-Escolar , Fator VIIIa/antagonistas & inibidores , Fator VIIIa/metabolismo , Fator VIIa/antagonistas & inibidores , Fator VIIa/metabolismo , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Análise Multivariada , Razão de Chances , Valor Preditivo dos Testes , Proteína C/antagonistas & inibidores , Proteína C/metabolismo , Fatores de Risco , alfa 1-Antitripsina/metabolismo
8.
Annu Rev Med ; 56: 63-77, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15660502

RESUMO

The development of new anticoagulants is expanding the list of drugs that can be used to prevent and treat venous and arterial thrombosis. New parenteral anticoagulants have been developed to overcome the limitations of heparin and low-molecular-weight heparin, whereas novel orally active anticoagulants have been designed to provide more streamlined therapy than vitamin K antagonists. This review identifies the molecular targets of new anticoagulants, describes the results of clinical trials, and provides clinical perspective on the opportunities for new anticoagulants.


Assuntos
Anticoagulantes/administração & dosagem , Drogas em Investigação/administração & dosagem , Trombose/tratamento farmacológico , Administração Oral , Antitrombina III/administração & dosagem , Ensaios Clínicos como Assunto , Fator VIIIa/antagonistas & inibidores , Fator VIIa/antagonistas & inibidores , Fator Va/antagonistas & inibidores , Humanos , Lipoproteínas/administração & dosagem , Trombina/antagonistas & inibidores , Trombose/sangue
9.
Biochemistry ; 42(38): 11316-25, 2003 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-14503882

RESUMO

The specific molecular target for direct heparin inhibition of factor X activation by intrinsic tenase (factor IXa-factor VIIIa) was investigated. Comparison of size-fractionated oligosaccharides demonstrated that an octasaccharide was sufficient to inhibit intrinsic tenase. Substitution of soluble dihexanoic phosphatidylserine (C6PS) for phospholipid (PL) vesicles demonstrated that inhibition by low-molecular weight heparin (LMWH) was independent of factor IXa-factor VIIIa membrane assembly. LMWH also inhibited factor X activation by the factor IXa-PL complex via a distinct mechanism that required longer oligosaccharides and was independent of substrate concentrations. The apparent affinity of LMWH for the factor IXa-PL complex was higher in the absence of factor VIIIa, suggesting that the cofactor adversely affected the interaction of heparin with factor IXa-phospholipid. LMWH did not interact directly with the active site, as it failed to inhibit chromogenic substrate cleavage by the factor IXa-PL complex. LMWH induced a modest decrease in factor IXa-factor VIIIa affinity [K(D(app))] on PL vesicles that did not account for the inhibition. In contrast, LMWH caused a substantial reduction in factor IXa-factor VIIIa affinity in the presence of C6PS that fully accounted for the inhibition. Factor IXa bound LMWH with significantly higher affinity than factor X by competition solution affinity analysis, and the K(D(app)) for the factor IXa-LMWH complex agreed with the K(I) for inhibition of the factor IXa-PL complex by LMWH. Thus, LMWH binds to an exosite on factor IXa that antagonizes cofactor activity without disrupting factor IXa-factor VIIIa assembly on the PL surface. This exosite may contribute to the clinical efficacy of heparin and represents a novel target for antithrombotic therapy.


Assuntos
Fator IXa/metabolismo , Heparina/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Sítios de Ligação , Ligação Competitiva , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Relação Dose-Resposta a Droga , Fator IXa/química , Fator VIIIa/antagonistas & inibidores , Fator VIIIa/química , Fator X/metabolismo , Heparina/análogos & derivados , Heparina/química , Heparina de Baixo Peso Molecular/química , Heparina de Baixo Peso Molecular/metabolismo , Heparina de Baixo Peso Molecular/farmacologia , Humanos , Cinética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Fosfolipídeos/química , Ressonância de Plasmônio de Superfície
10.
Semin Vasc Med ; 3(2): 193-8, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-15199482

RESUMO

This article focuses on the role of the tissue factor (TF)-thrombin pathway in cardiac ischemia-reperfusion (I/R) injury. We and others have used rabbit models of cardiac I/R injury to show that anti-TF therapy prevents the transient decrease in regional myocardial blood flow, reduces platelet and fibrin(ogen) accumulation, and reduces infarct size. At present, the mechanism by which TF-initiated coagulation contributes to myocardial injury is not established. Inhibition of TF may decrease intravascular fibrin deposition and thrombosis. However, immunohistochemical studies demonstrated that fibrin deposition was predominantly within the myocardium and depletion of fibrinogen did not reduce infarct size. In contrast, inhibition of thrombin reduced infarct size to a similar extent as anti-TF therapy. We propose that the TF-thrombin pathway may contribute to myocardial injury by an additional mechanism that is not dependent on fibrin deposition but involves activation of protease activated receptor 1 (PAR-1) on vascular endothelial cells and cardiac myocytes. Anti-TF therapy would inhibit both thrombin-dependent fibrin deposition and thrombin-dependent PAR-1 signaling.


Assuntos
Traumatismo por Reperfusão Miocárdica/fisiopatologia , Trombina/fisiologia , Tromboplastina/fisiologia , Animais , Coagulação Sanguínea/fisiologia , Fator VIIIa/antagonistas & inibidores , Fibrinogênio/fisiologia , Humanos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Miocárdio/metabolismo , Coelhos , Receptor PAR-1/fisiologia , Trombina/antagonistas & inibidores , Tromboplastina/antagonistas & inibidores
11.
Semin Vasc Med ; 3(2): 205-14, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-15199484

RESUMO

It is generally accepted that the initial event in coagulation and intravascular thrombus formation is the exposure of cell-surface protein, such as tissue factor (TF). TF is exposed to the flowing blood as a consequence of vascular injury induced, for instance, by PTCA, or by spontaneous rupture of an atherosclerotic plaque. Expression of TF may also be induced in monocytes and endothelial cells in several conditions such as sepsis and cancer, causing a more generalized activation of clotting. In addition to its essential role in hemostasis, TF may be also implicated in several pathophysiological processes, such as intracellular signaling, cell proliferation, and inflammation. For all these reasons, TF has been the subject of intense research focus. Many experimental studies have demonstrated that inhibition of TF:factor VIIa procoagulant activity is a powerful inhibitor of in vivo thrombosis and that this approach usually results in a less-pronounced bleeding tendency compared with other "more classical" antithrombotic interventions. Alternative approaches may be represented by antibodies directed against TF, by transfection of the arterial wall with natural inhibitors of the TF:factor VIIa complex, such as the TF pathway inhibitor, or with catalytic RNA (ribozyme), which could inhibit the expression of the TF protein by the disruption of cellular TF mRNA. All these approaches seem particularly attractive because they may result in complete inhibition of local thrombosis without incurring potentially harmful systemic effects. Further studies are warranted to determine the efficacy and safety of such approaches in patients.


Assuntos
Tromboplastina/antagonistas & inibidores , Trombose/fisiopatologia , Animais , Arteriosclerose/fisiopatologia , Coagulação Sanguínea/fisiologia , Fator VIIIa/antagonistas & inibidores , Fator VIIIa/fisiologia , Fibrinolíticos/uso terapêutico , Humanos , Lipoproteínas/farmacologia , Lipoproteínas/uso terapêutico , Transdução de Sinais , Tromboplastina/fisiologia , Trombose/prevenção & controle
12.
Biochemistry ; 40(29): 8508-13, 2001 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-11456488

RESUMO

A monoclonal IgG isolated from a patient with multiple myeloma has been shown to bind to exosite II of thrombin, prolong both the thrombin time and the activated partial thromboplastin time (aPTT) when added to normal plasma, and alter the kinetics of hydrolysis of synthetic peptide substrates. Although the IgG does not affect cleavage of fibrinogen by thrombin, it increases the rate of inhibition of thrombin by purified antithrombin approximately 3-fold. Experiments with plasma immunodepleted of antithrombin or heparin cofactor II confirm that prolongation of the thrombin time requires antithrombin. By contrast, prolongation of the aPTT requires neither antithrombin nor heparin cofactor II. The IgG delays clotting of plasma initiated by purified factor IXa but has much less of an effect on clotting initiated by factor Xa. In a purified system, the IgG decreases the rate of activation of factor VIII by thrombin. These studies indicate that binding of a monoclonal IgG to exosite II prolongs the thrombin time indirectly by accelerating the thrombin-antithrombin reaction and may prolong the aPTT by interfering with activation of factor VIII, thereby diminishing the catalytic activity of the factor IXa/VIIIa complex.


Assuntos
Anticorpos Monoclonais/metabolismo , Anticoagulantes/metabolismo , Antitrombinas/metabolismo , Sítios de Ligação de Anticorpos , Trombina/imunologia , Trombina/metabolismo , Ânions/metabolismo , Relação Dose-Resposta Imunológica , Fator IXa/antagonistas & inibidores , Fator IXa/metabolismo , Fator VIII/antagonistas & inibidores , Fator VIII/metabolismo , Fator VIIIa/antagonistas & inibidores , Fator VIIIa/metabolismo , Fibrinogênio/isolamento & purificação , Fibrinogênio/metabolismo , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Imunoglobulina G/metabolismo , Tempo de Tromboplastina Parcial , Tempo de Trombina
13.
Thromb Haemost ; 85(5): 761-5, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11372664

RESUMO

Activated protein C (APC) is an important inactivator of coagulation factors Va and VIIIa. In the inactivation of factors Va and VIIIa, protein S serves as a cofactor to APC. Protein S can bind to C4b-binding protein (C4BP), and thereby loses its cofactor activity to APC. By modulating free protein S levels, C4BP is an important regulator of protein S cofactor activity. In the factor VIIIa inactivation, protein S and factor V act as synergistic cofactors to APC. We investigated the effect of C4BP on both the factor V-independent and factor V-dependent cofactor activity of protein S in the factor VIIIa inactivation using a purified system. Protein S increased the APC-mediated inactivation of factor VIIIa to 60% and in synergy with protein S, factor V at equimolar concentrations increased this effect further to 90%. The protein S/factor V synergistic effect was inhibited by preincubation of protein S and factor V with a four-fold molar excess of C4BP. However, C4BP did not inhibit the factor V-independent protein S cofactor activity in the purified system whereas it inhibited the cofactor activity in plasma. We conclude that C4BP-bound protein S retains its cofactor activity to APC in the factor VIIIa inactivation.


Assuntos
Fator VIIIa/antagonistas & inibidores , Fator V/farmacologia , Integrina alfaXbeta2/farmacologia , Proteína C/farmacologia , Proteína S/antagonistas & inibidores , Anticoagulantes/antagonistas & inibidores , Anticoagulantes/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Fator VIIIa/metabolismo , Humanos , Integrina alfaXbeta2/metabolismo , Cinética , Membranas Artificiais , Fosfolipídeos/metabolismo , Ligação Proteica , Proteína C/metabolismo , Proteína S/efeitos dos fármacos , Proteína S/metabolismo , Proteína S/farmacologia
14.
Biochemistry ; 40(16): 4980-9, 2001 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-11305914

RESUMO

Phosphorothioate oligonucleotides (PS ODNs) prolong the activated partial thromboplastin time in human plasma by inhibition of intrinsic tenase (factor IXa-factor VIIIa) activity. This inhibition was characterized using ISIS 2302, a 20-mer antisense PS ODN. ISIS 2302 demonstrated hyperbolic, mixed-type inhibition of factor X activation by the intrinsic tenase complex. The decrease in V(max(app)) was analyzed by examining complex assembly, cofactor stability, and protease catalysis. ISIS 2302 did not inhibit factor X activation by the factor IXa-phospholipid complex, or significantly affect factor VIII-phospholipid affinity. Inhibitory concentrations of ISIS 2302 modestly decreased the affinity of factor IXa-factor VIIIa binding in the presence of phospholipid (K(D) = 11.5 vs 4.8 nM). This effect was insufficient to explain the reduction in V(max(app)). ISIS 2302 did not affect the in vitro half-life of factor VIIIa, suggesting it did not destabilize cofactor activity. In the presence of 30% ethylene glycol, the level of factor X activation by the factor IXa-phospholipid complex increased 3-fold, and the level of chromogenic substrate cleavage by factor IXa increased more than 50-fold. ISIS 2302 demonstrated partial inhibition of factor X activation by the factor IXa-phospholipid complex, and chromogenic substrate cleavage by factor IXa, only in the presence of ethylene glycol. Like the intact enzyme complex, ISIS 2302 demonstrated hyperbolic, mixed-type inhibition of chromogenic substrate cleavage by factor IXa (K(I) = 88 nM). Equilibrium binding studies with fluorescein-labeled ISIS 2302 demonstrated a similar affinity (K(D) = 92 nM) for the PS ODN-factor IX interaction. These results suggest that PS ODNs bind to an exosite on factor IXa, modulating catalytic activity of the intrinsic tenase complex.


Assuntos
Cisteína Endopeptidases/metabolismo , Fator IXa/antagonistas & inibidores , Fator VIIIa/antagonistas & inibidores , Proteínas de Neoplasias , Inibidores de Serina Proteinase/farmacologia , Tionucleotídeos/farmacologia , Regulação Alostérica , Sítios de Ligação/efeitos dos fármacos , Ligação Competitiva , Catálise , Etilenoglicol/farmacologia , Fator IXa/metabolismo , Fator VIIIa/metabolismo , Meia-Vida , Humanos , Substâncias Macromoleculares , Oligodesoxirribonucleotídeos Antissenso/metabolismo , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Fosfolipídeos/antagonistas & inibidores , Fosfolipídeos/metabolismo , Oligonucleotídeos Fosforotioatos , Inibidores de Serina Proteinase/classificação , Tionucleotídeos/metabolismo
15.
J Exp Med ; 190(1): 91-9, 1999 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-10429673

RESUMO

Agents that restore vascular patency in stroke also increase the risk of intracerebral hemorrhage (ICH). As Factor IXa is a key intermediary in the intrinsic pathway of coagulation, targeted inhibition of Factor IXa-dependent coagulation might inhibit microvascular thrombosis in stroke without impairing extrinsic hemostatic mechanisms that limit ICH. A competitive inhibitor of native Factor IXa for assembly into the intrinsic Factor X activation complex, Factor IXai, was prepared by covalent modification of the Factor IXa active site. In a modified cephalin clotting time assay, in vivo administration of Factor IXai caused a dose-dependent increase in time to clot formation (3.6-fold increase at the 300 micrograms/kg dose compared with vehicle-treated control animals, P < 0.05). Mice given Factor IXai and subjected to middle cerebral artery occlusion and reperfusion demonstrated reduced microvascular fibrin accumulation by immunoblotting and immunostaining, reduced 111In-labeled platelet deposition (42% decrease, P < 0.05), increased cerebral perfusion (2.6-fold increase in ipsilateral blood flow by laser doppler, P < 0.05), and smaller cerebral infarcts than vehicle-treated controls (70% reduction, P < 0.05) based on triphenyl tetrazolium chloride staining of serial cerebral sections. At therapeutically effective doses, Factor IXai was not associated with increased ICH, as opposed to tissue plasminogen activator (tPA) or heparin, both of which significantly increased ICH. Factor IXai was cerebroprotective even when given after the onset of stroke, indicating that microvascular thrombosis continues to evolve (and may be inhibited) even after primary occlusion of a major cerebrovascular tributary.


Assuntos
Hemorragia Cerebral/prevenção & controle , Ataque Isquêmico Transitório/fisiopatologia , Animais , Coagulação Sanguínea/efeitos dos fármacos , Fator IXa/antagonistas & inibidores , Fator VIIIa/antagonistas & inibidores , Fator X/antagonistas & inibidores , Hemostasia/fisiologia , Ataque Isquêmico Transitório/patologia , Camundongos , Grau de Desobstrução Vascular/efeitos dos fármacos
16.
Blood ; 92(5): 1617-25, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9716589

RESUMO

Systemic administration of ISIS 2302, a 20-mer antisense phosphorothioate oligonucleotide targeting human intercellular adhesion molecule-1 mRNA, causes prolongation of plasma clotting times in both monkey and human studies. The anticoagulant effects of ISIS 2302 were investigated with both in vitro coagulation assays in human plasma and purified enzyme systems. At high oligonucleotide plasma concentrations (>100 microgram/mL), prolongation of the prothrombin and thrombin times was observed. In a thrombin time assay using purified components, high concentrations of ISIS 2302 inhibited thrombin clotting activity both by stimulating inhibition by heparin cofactor II and directly competing with fibrinogen for binding to anion binding exosite I. In contrast, low concentrations of ISIS 2302 (<100 microgram/mL) showed a selective, linear prolongation of the activated partial thromboplastin time (PTT). The rate limiting effect of 50 microgram/mL ISIS 2302, which prolonged the PTT to 1.5 times control, was identified by sequential modification of the clotting assay. Delaying addition of oligonucleotide until after contact activation failed to correct prolongation of the PTT. The calcium-dependent steps of the intrinsic pathway were individually assessed by adding sufficient activated coagulation factor to correct the PTT in plasma deficient in that specific factor. Addition of factor XIa, IXa, VIIIa, or Va failed to correct the PTT in the presence of ISIS 2302. In contrast, 0.2 nmol/L factor Xa corrected prolongation of the PTT in factor X-deficient plasma with or without oligonucleotide present. ISIS 2302 (50 microgram/mL) did not prolong a modified Russel viper venom time, suggesting no significant inhibition of prothrombinase. Thus, 50 microgram/mL ISIS 2302 prolonged the PTT by selectively inhibiting intrinsic tenase activity. ISIS 2302 showed partial inhibition of intrinsic tenase activity (to approximately 35% of control) at clinically relevant oligonucleotide concentrations in a chromogenic assay. This activity was oligonucleotide sequence-independent but required the phosphorothioate backbone, suggesting that inhibition of intrinsic tenase is a general property of this class of oligonucleotides. These results are relevant to both the therapeutic use of phosphorothioate oligonucleotides and the potential design of inhibitors of the intrinsic tenase complex, a novel target for anticoagulation.


Assuntos
Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fator IXa/antagonistas & inibidores , Fator VIIIa/antagonistas & inibidores , Oligodesoxirribonucleotídeos Antissenso , Oligonucleotídeos Antissenso/farmacologia , Tionucleotídeos/farmacologia , Animais , Antitrombina III/antagonistas & inibidores , Ligação Competitiva , Cálcio/sangue , Fator IXa/metabolismo , Fator VIIIa/metabolismo , Fibrinogênio/metabolismo , Cofator II da Heparina/antagonistas & inibidores , Humanos , Tempo de Tromboplastina Parcial , Fosfolipídeos/sangue , Oligonucleotídeos Fosforotioatos , Tempo de Protrombina , Coelhos , Tempo de Trombina
17.
Proc Natl Acad Sci U S A ; 94(22): 11851-6, 1997 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-9342326

RESUMO

Individuals with hemophilia A require frequent infusion of preparations of coagulation factor VIII. The activity of factor VIII (FVIII) as a cofactor for factor IXa in the coagulation cascade is limited by its instability after activation by thrombin. Activation of FVIII occurs through proteolytic cleavage and generates an unstable FVIII heterotrimer that is subject to rapid dissociation of its subunits. In addition, further proteolytic cleavage by thrombin, factor Xa, factor IXa, and activated protein C can lead to inactivation. We have engineered and characterized a FVIII protein, IR8, that has enhanced in vitro stability of FVIII activity due to resistance to subunit dissociation and proteolytic inactivation. FVIII was genetically engineered by deletion of residues 794-1689 so that the A2 domain is covalently attached to the light chain. Missense mutations at thrombin and activated protein C inactivation cleavage sites provided resistance to proteolysis, resulting in a single-chain protein that has maximal activity after a single cleavage after arginine-372. The specific activity of partially purified protein produced in transfected COS-1 monkey cells was 5-fold higher than wild-type (WT) FVIII. Whereas WT FVIII was inactivated by thrombin after 10 min in vitro, IR8 still retained 38% of peak activity after 4 hr. Whereas binding of IR8 to von Willebrand factor (vWF) was reduced 10-fold compared with WT FVIII, in the presence of an anti-light chain antibody, ESH8, binding of IR8 to vWF increased 5-fold. These results demonstrate that residues 1690-2332 of FVIII are sufficient to support high-affinity vWF binding. Whereas ESH8 inhibited WT factor VIII activity, IR8 retained its activity in the presence of ESH8. We propose that resistance to A2 subunit dissociation abrogates inhibition by the ESH8 antibody. The stable FVIIIa described here provides the opportunity to study the activated form of this critical coagulation factor and demonstrates that proteins can be improved by rationale design through genetic engineering technology.


Assuntos
Fator VIIIa/genética , Fator VIIIa/metabolismo , Engenharia de Proteínas/métodos , Ativação Enzimática , Fator VIIIa/antagonistas & inibidores , Ligação Proteica , Proteína C/metabolismo , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Trombina/metabolismo , Fator de von Willebrand/metabolismo
18.
Thromb Haemost ; 78(3): 1030-6, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9308749

RESUMO

Human factor V has been shown not only to be a precursor to procoagulant factor Va but also to express anticoagulant properties. Thus, factor V was recently found to potentiate the effect of protein S as cofactor to activated protein C (APC) in the inactivation of the factor VIIIa-factor IXa complex. The purpose of this study was to determine whether the APC-cofactor function of factor V was also expressed in the bovine protein C system and to elucidate the molecular background for the species specificity of APC. For this purpose, the effects of protein S and factor V on APC-mediated inactivation of factor VIIIa were studied using purified APC, protein S and factor V of human and bovine origin. The factor VIIIa investigated here was part of a Xase complex (i.e. factor IXa, factor VIIIa, phospholipid and calcium) and the APC-mediated inhibition of factor VIIIa was monitored by the ability of the Xase complex to activate factor X. Synergistic APC-cofactor function of factor V and protein S was demonstrated in the bovine system. The effect of bovine APC was potentiated by bovine protein S but not by human protein S, whereas both human or bovine protein S stimulated the function of human APC. Factor V did not express species specificity in its APC-cofactor activity even though bovine factor V was more potent than its human counterpart. Recombinant human/bovine protein S chimeras were used to demonstrate that the thrombin sensitive region and first epidermal growth factor-like module of protein S determine the species specificity of the APC-protein S interaction. In conclusion, both human and bovine factor V were found to express APC-cofactor activity which depends on the presence of protein S. The species specificity of APC was shown to be caused by the interaction between APC and protein S.


Assuntos
Fatores de Coagulação Sanguínea/antagonistas & inibidores , Coagulação Sanguínea/fisiologia , Cisteína Endopeptidases/metabolismo , Fator IXa/antagonistas & inibidores , Fator VIIIa/antagonistas & inibidores , Fator V/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteína C/metabolismo , Proteína S/farmacologia , Animais , Sítios de Ligação , Bovinos , Sinergismo Farmacológico , Humanos , Especificidade da Espécie
19.
J Clin Invest ; 93(6): 2497-504, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8200986

RESUMO

Human inhibitory alloantibodies and autoantibodies to Factor VIII (FVIII) are usually directed toward the A2 and/or C2 domains of the FVIII molecule. Anti-C2 antibodies block the binding of FVIII to phospholipid, but the mechanism of action of anti-A2 antibodies is not known. We investigated the properties of a patient autoantibody, RC, and a monoclonal antibody, 413, that bind to the region which contains the epitopes of all anti-A2 alloantibodies or autoantibodies studied to date. mAb 413 and RC were noncompetitive inhibitors of a model intrinsic Factor X activation complex (intrinsic FXase) consisting of Factor IXa, activated FVIII (FVIIIa), and synthetic phospholipid vesicles, since they decreased the Vmax of intrinsic FXase by > 95% at saturating concentrations without altering the Km. This indicates that RC and mAb 413 either block the binding of FVIIIa to FIXa or phospholipid or interfere with the catalytic function of fully assembled intrinsic FXase, but they do not inhibit the binding of the substrate Factor X. mAb 413 did not inhibit the increase in fluorescence anisotropy that results from the binding of Factor VIIIa to fluorescein-5-maleimidyl-D-phenylalanyl-prolyl-arginyl-FIXa (Fl-M-FPR-FIXa) on phospholipid vesicles in the absence of Factor X, indicating it does not inhibit assembly of intrinsic FXase. Addition of Factor X to Fl-M-FPR-FIXa, FVIIIa, and phospholipid vesicles produced a further increase in fluorescence anisotropy and a decrease in fluorescence intensity. This effect was blocked completely by mAb 413. We conclude that anti-A2 antibodies inhibit FVIIIa function by blocking the conversion of intrinsic FXase/FX complex to the transition state, rather than by interfering with formation of the ground state Michaelis complex.


Assuntos
Anticorpos Monoclonais/imunologia , Autoanticorpos/imunologia , Fator VIIIa/antagonistas & inibidores , Animais , Fator VIIIa/metabolismo , Fator X/farmacologia , Polarização de Fluorescência , Humanos , Camundongos
20.
J Biol Chem ; 266(36): 24514-9, 1991 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-1761551

RESUMO

Activated protein C (APC) exerts its physiologic anticoagulant role by proteolytic inactivation of the blood coagulation cofactors Va and VIIIa. To identify the regions on the surface that mediate anticoagulant activity, 26 synthetic peptides were prepared representing 90% of the human protein C heavy chain primary structure and tested for their ability to inhibit APC anticoagulant activity. Peptide-(390-404) specifically inhibited APC activity in activated partial thromboplastin time and Xa-1-stage coagulation assays in normal, in protein S-depleted and Factor VIII-deficient plasma with 50% inhibition at 5 microM peptide. Polyclonal antibodies raised against this peptide and immunoaffinity-purified on a protein C-Sepharose column inhibited APC anticoagulant activity in activated partial thromboplastin time and Xa-1-stage assays in normal, protein S-depleted, and Factor VIII-deficient plasma with half-maximal inhibition at 30 nM anti-(390-404) antibody. Neither the peptide-(390-404) nor the anti-(390-404) antibodies inhibited APC amidolytic activity or the reaction of APC with recombinant [Arg358] alpha 1-antitrypsin. Furthermore, in a purified system, peptide-(390-404) inhibited APC-catalyzed inactivation of Factor Va in the presence as well as in the absence of phospholipids with 50% inhibition at 4 microM peptide. These data suggest that the region containing residues 390-404 in APC is essential for anticoagulant activity and is available to interact with antibodies or with other proteins such as the macromolecular substrates Factors Va or VIIIa.


Assuntos
Coagulação Sanguínea , Proteína C/fisiologia , Sequência de Aminoácidos , Catálise , Fator VIIIa/antagonistas & inibidores , Fator Va/antagonistas & inibidores , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/fisiologia , Proteína C/genética , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA