Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Mol Biol ; 431(5): 885-903, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30653992

RESUMO

Proprotein convertase subtilisin/kexin 9 (PCSK9) is a key regulator of lipid metabolism by degrading liver LDL receptors. Structural studies have provided molecular details of PCSK9 function. However, the N-terminal acidic stretch of the PCSK9 prodomain (Q31-T60) has eluded structural investigation, since it is in a disordered state. The interest in this region is intensified by the presence of human missense mutations associated with low and high LDL-c levels (E32K, D35Y, and R46L, respectively), as well as two posttranslationally modified sites, sulfated Y38 and phosphorylated S47. Herein we show that a segment within this region undergoes disorder-to-order transition. Experiments with acidic stretch-derived peptides demonstrated that the folding is centered at the segment Y38-L45, which adopts an α-helix as determined by NMR analysis of free peptides and by X-ray crystallography of peptides in complex with antibody 6E2 (Ab6E2). In the Fab6E2-peptide complexes, the structured region features a central 2 1/4-turn α-helix and encompasses up to 2/3 of the length of the acidic stretch, including the missense mutations and posttranslationally modified sites. Experiments with helix-breaking proline substitutions in peptides and in PCSK9 protein indicated that Ab6E2 specifically recognizes the helical conformation of the acidic stretch. Therefore, the observed quantitative binding of Ab6E2 to native PCSK9 from various cell lines suggests that the disorder-to-order transition is a true feature of PCSK9 and not limited to peptides. Because the helix provides a constrained spatial orientation of the missense mutations and the posttranslationally modified residues, it is probable that their biological functions take place in the context of an ordered conformational state.


Assuntos
Pró-Proteína Convertase 9/genética , Sequência de Aminoácidos , Linhagem Celular Tumoral , Cristalografia por Raios X/métodos , Células Hep G2 , Humanos , Mutação de Sentido Incorreto/genética , Peptídeos/genética , Processamento de Proteína Pós-Traducional/genética , Receptores de LDL/genética , Serina Endopeptidases/genética
2.
J Thromb Haemost ; 13(9): 1676-82, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26176663

RESUMO

BACKGROUND: We aimed to evaluate the mechanisms underlying the effects of red blood cells (RBCs) on the reactivity of monocytes to lipopolysaccharide (LPS) stimulation. METHODS: Measurements of tissue factor (TF) antigen and activity were performed on freshly isolated white blood cells (WBCs)/platelets resuspended in heparinized plasma, as well as cultured monocytic cells. RESULTS: In a dose-dependent manner, RBCs significantly enhanced LPS-induced TF activity and antigen levels in blood monocytes; potentiation of TF activity by both human and murine RBCs did not require the presence of neutrophils and/or platelets. We also measured the levels of monocyte chemotactic protein-1 (MCP-1), the key proinflammatory chemokine that binds to duffy antigen receptor for chemokines (DARC) on RBC surface, in plasma and RBC lysates after the incubation of RBCs with WBC/platelets; at the concentrations corresponding to normal blood counts, RBCs exerted a significant influence on the free plasma levels of MCP-1, with about two-thirds of detectable MCP-1 post-LPS stimulation being associated with RBCs. Critically, DARC-deficient murine RBCs failed to enhance LPS-induced TF activity, confirming the mechanistic significance of RBC-DARC. CONCLUSIONS: Our study reports a novel mechanism by which RBCs promote procoagulant and proinflammatory sequelae of WBC exposure to LPS, likely mediated by RBC-DARC in the microenvironment(s) that bring monocytes and RBCs in close proximity.


Assuntos
Coagulação Sanguínea , Quimiocina CCL2 , Sistema do Grupo Sanguíneo Duffy , Eritrócitos , Inflamação , Monócitos , Receptores de Superfície Celular , Tromboplastina , Adulto , Animais , Humanos , Camundongos , Coagulação Sanguínea/fisiologia , Linhagem Celular , Quimiocina CCL2/biossíntese , Quimiocina CCL2/sangue , Quimiocina CCL2/genética , Sistema do Grupo Sanguíneo Duffy/sangue , Sistema do Grupo Sanguíneo Duffy/imunologia , Endotoxemia/sangue , Endotoxemia/imunologia , Eritrócitos/imunologia , Regulação da Expressão Gênica , Inflamação/sangue , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/toxicidade , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/metabolismo , Receptores de Superfície Celular/sangue , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/imunologia , RNA Mensageiro/biossíntese , RNA Mensageiro/sangue , Tromboplastina/biossíntese , Tromboplastina/genética
3.
J Thromb Haemost ; 7(7): 1092-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19422446

RESUMO

BACKGROUND: Tissue factor (TF) is present in blood in various forms, including small membrane vesicles called microparticles (MPs). Elevated levels of these MPs appear to play a role in the pathogenesis of thrombosis in a variety of diseases, including sepsis. OBJECTIVE: Measure levels of MP TF activity and activation of coagulation in control and endotoxemic mice. MATERIALS AND METHODS: MPs were prepared from plasma by centrifugation. The procoagulant activity (PCA) of MPs was measured using a two-stage chromogenic assay. We also measured levels of thrombin-antithrombin and the number of MPs. RESULTS: Lipopolysaccharide (LPS) increased MP PCA in wild-type mice; this PCA was significantly reduced by an anti-mouse TF antibody (1H1) but not with an anti-human TF antibody (HTF-1). Conversely, in mice expressing only human TF, MP PCA was inhibited by HTF-1 but not 1H1. MPs from wild-type mice had 6-fold higher levels of PCA using mouse factor (F)VIIa compared with human FVIIa, which is consistent with reported species-specific differences in FVIIa. Mice expressing low levels of human TF had significantly lower levels of MP TF activity and TAT than mice expressing high levels of human TF; however, there were similar levels of phosphatidylserine (PS)-positive MPs. Importantly, levels of MP TF activity in wild-type mice correlated with levels of TAT but not with PS-positive MPs in endotoxemic mice. CONCLUSION: These results suggest that the levels of TF-positive MPs can be used as a biomarker for evaluating the risk of disseminated intravascular coagulation in endotoxemia.


Assuntos
Coagulação Sanguínea , Endotoxemia/fisiopatologia , Tromboplastina/metabolismo , Animais , Ensaio de Imunoadsorção Enzimática , Humanos , Camundongos
5.
J Thromb Haemost ; 1(5): 889-95, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12871353

RESUMO

Antibody mediated inhibition of tissue factor (TF) function reduces thrombus size in ex vivo perfusion of human blood over a TF-free surface at venous shear rates suggesting that TF might be involved in the mechanism of deep vein thrombosis. Moreover, TF-bearing monocytes and polymorphonuclear (PMN) leukocytes were identified in human ex vivo formed thrombi and in circulating blood. To understand the role of TF in thrombus growth, we applied a rabbit venous thrombosis model in which a collagen-coated thread was installed within the jugular vein or within a silicon vein shunt. The effect of an inhibitory monoclonal antirabbit TF antibody (AP-1) or Napsagatran, a specific inhibitor of thrombin, was quantified by continuously monitoring 125I-fibrinogen incorporation into the growing thrombi. The antithrombotic effect obtained with the anti-TF antibody was comparable to the effect observed with the thrombin inhibitor napsagatran suggesting that in this animal model the thrombus propagation is highly TF dependent. Immunostaining revealed that TF was mostly associated with leukocytes within the thrombi formed in the jugular vein or in the silicon vein shunt. Ex vivo perfusion experiments over collagen-coated coverslips demonstrated the presence of TF-bearing PMN leukocytes in circulating blood. The results suggest that in rabbits venous thrombus growth is mediated by clot-bound TF and that blocking the TF activity can inhibit thrombus propagation.


Assuntos
Tromboplastina/fisiologia , Trombose Venosa/etiologia , Animais , Anticorpos Monoclonais/farmacologia , Prótese Vascular , Fibrinogênio/metabolismo , Imuno-Histoquímica , Veias Jugulares , Leucócitos/química , Naftalenos/farmacologia , Piperidinas/farmacologia , Coelhos , Tromboplastina/análise , Tromboplastina/antagonistas & inibidores , Trombose Venosa/patologia
6.
J Mol Biol ; 313(1): 83-97, 2001 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-11601848

RESUMO

The outstanding importance of the antigen-antibody recognition process for the survival and defence strategy of higher organisms is in sharp contrast to the limited high resolution structural data available on antibody-antigen pairs with antigenic proteins. The limitation is the most severe for structural data not restricted to the antigen-antibody complex but extending to the uncomplexed antigen and antibody. We report the crystal structure of the complex between tissue factor (TF) and the humanized Fab fragment D3h44 at a resolution of 1.85 A together with the structure of uncomplexed D3h44 at the same resolution. In conjunction with the previously reported 1.7 A crystal structure of uncomplexed TF, a unique opportunity is generated to explore details of the recognition process. The TF.D3h44 interface is characterised by a high number of polar interactions, including as may as 46 solvent molecules. Conformational changes upon complex formation are very small and almost exclusively limited to the reorientation of side-chains. The binding epitope is in complete agreement with earlier mutagenesis experiments. A revaluation of two other antibody-antigen pairs reported at similar resolutions, shows that all these complexes are very similar with respect to the solvation of the interface, the number of solvent positions conserved in the uncomplexed and complexed proteins and the number of water molecules expelled from the surface and replaced by hydrophilic atoms from the binding partner upon complex formation. A strategy is proposed on how to exploit this high resolution structural data to guide the affinity maturation of humanised antibodies.


Assuntos
Sítios de Ligação de Anticorpos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/imunologia , Tromboplastina/química , Tromboplastina/imunologia , Água/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Solventes , Tromboplastina/metabolismo , Água/química
8.
Structure ; 9(7): 627-36, 2001 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-11470437

RESUMO

BACKGROUND: Coagulation factor VIIa (FVIIa) contains a Trypsin-like serine protease domain and initiates the cascade of proteolytic events leading to Thrombin activation and blood clot formation. Vascular injury allows formation of the complex between circulating FVIIa and its cell surface bound obligate cofactor, Tissue Factor (TF). Circulating FVIIa is nominally activated but retains zymogen-like character and requires TF in order to complete the zymogen-to-enzyme transition. The manner in which TF exerts this effect is unclear. The structure of TF/FVIIa is known. Knowledge of the zymogen structure is helpful for understanding the activation transition in this system. RESULTS: The 2 A resolution crystal structure of a zymogen form of FVII comprising the EGF2 and protease domains is revealed in a complex with the exosite binding inhibitory peptide A-183 and a vacant active site. The activation domain, which includes the N terminus, differs in ways beyond those that are expected for zymogens in the Trypsin family. There are large differences in the TF binding region. An unprecedented 3 residue shift in registration between beta strands B2 and A2 in the C-terminal beta barrel and hydrogen bonds involving Glu154 provide new insight into conformational changes accompanying zymogen activation, TF binding, and enzymatic competence. CONCLUSIONS: TF-mediated allosteric control of the activity of FVIIa can be rationalized. The reregistering beta strand connects the TF binding region and the N-terminal region. The zymogen registration allows H bonds that prevent the N terminus from attaining a key salt bridge with the active site. TF binding may influence an equilibrium by selecting the enzymatically competent registration.


Assuntos
Fator VII/química , Fator VII/metabolismo , Ácido Aspártico/química , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Precursores Enzimáticos/química , Fator VII/genética , Ácido Glutâmico/química , Isoleucina/química , Modelos Moleculares , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tromboplastina/metabolismo
9.
Thromb Haemost ; 85(3): 379-89, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11307801

RESUMO

Blocking the cofactor function of human tissue factor may be beneficial in various coagulation-mediated diseases. The murine antibody D3 binds to the membrane proximal substrate interaction region of human tissue factor and blocks tissue factor function even in the presence of bound factor VIIa. The cloned murine D3 antibody was humanized and affinity matured by exchanging amino acids in the complementarity determining regions as well as in the antibody framework. The humanized antibody, D3H44, bound to tissue factor with a 100-fold increased affinity (KD 0.1 nM) as compared to the original murine and chimeric versions. Depending on the particular disease, different pharmacokinetic properties of the antibody may be required and, therefore, several antibody variants-- F(ab), F(ab')2, IgG2, IgG4 and IgG4b-were generated. In vitro, the humanized D3 antibodies displayed potent inhibition of plasma clotting and tissue factor: factor VIIa-mediated activation of factors IX and X (e.g. D3H44-F(ab')2, IC50(F.X) 47 pM). In addition, D3H44-F(ab')2 completely prevented fibrin deposition in a human ex vivo thrombosis model under venous blood flow conditions (IC50 37 nM). The humanized D3 antibodies may be utilized for treatment of cardiovascular diseases which involve tissue factor activity, e.g. acute coronary syndrome and venous thrombosis.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Afinidade de Anticorpos/imunologia , Tromboplastina/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Testes de Coagulação Sanguínea , Clonagem Molecular , Fator IX/efeitos dos fármacos , Fator IX/metabolismo , Fator X/efeitos dos fármacos , Fator X/metabolismo , Fibrinolíticos/farmacologia , Fibrinolíticos/uso terapêutico , Humanos , Fragmentos de Imunoglobulinas/genética , Camundongos , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Alinhamento de Sequência
10.
Thromb Haemost ; 85(3): 475-81, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11307818

RESUMO

The substrate recognition region of tissue factor contains two residues, Lys165 and Lys166, which are important for macromolecular substrate activation by the tissue factor:factor VIIa complex. Replacement of these two residues with alanine in a soluble version of human tissue factor resulted in a mutant, hTFAA, which can bind factor VIIa but forms an enzymatically inactive complex. We found that hTFAA inhibits the activity of guinea pig factor VIIa, allowing us to evaluate hTFAA's effects on thrombosis and hemostasis in a guinea pig model of recurrent arterial thrombosis. In addition to heparin, the effects of hTFAA were compared to active site inhibited factor IXa (F.IXai) and factor Xa (F.Xai). We found that hTFAA, F.IXai and F.Xai were potent antithrombotics and may possess a decreased risk of hemorrhage when compared to unfractionated heparin. When administered at a dose that inhibited thrombosis by about 90%, hTFAA neither affected cuticle bleeding nor the activated partial thromboplastin time, and had only a modest effect on the prothrombin time. At equi-efficacious doses, F.IXai, F.Xai and heparin prolonged bleeding times by 20% (p >0.5), 50% (p <0.05) and 100% (p <0.01), respectively. In summary, our study demonstrates that, unlike heparin, specific inhibitors of factors VIIa, IXa and Xa can produce antithrombotic effects without or with only minimally disturbing normal hemostasis. The results further suggest that factor VIIa and factor IXa are especially promising targets for antithrombotic drug development.


Assuntos
Tromboplastina/genética , Trombose/prevenção & controle , Substituição de Aminoácidos , Animais , Arteriopatias Oclusivas/tratamento farmacológico , Arteriopatias Oclusivas/etiologia , Arteriopatias Oclusivas/prevenção & controle , Tempo de Sangramento , Trombose das Artérias Carótidas/tratamento farmacológico , Trombose das Artérias Carótidas/etiologia , Trombose das Artérias Carótidas/prevenção & controle , Domínio Catalítico , Modelos Animais de Doenças , Fator IXa/antagonistas & inibidores , Fator IXa/farmacologia , Fator VIIa/efeitos dos fármacos , Fator VIIa/metabolismo , Fator Xa/farmacologia , Inibidores do Fator Xa , Fibrinolíticos/administração & dosagem , Fibrinolíticos/farmacologia , Cobaias , Heparina/administração & dosagem , Heparina/farmacologia , Humanos , Solubilidade , Tromboplastina/administração & dosagem , Tromboplastina/farmacologia , Trombose/tratamento farmacológico , Trombose/etiologia
11.
Biochemistry ; 40(3): 675-82, 2001 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-11170384

RESUMO

Tissue factor is the cell membrane-anchored cofactor for factor VIIa and triggers the coagulation reactions. The initial step is the conversion of factor VII to factor VIIa which, in vitro, is efficiently catalyzed by low concentrations of factor Xa. To identify the tissue factor region that interacts with the activator factor Xa during this process, we evaluated a panel of soluble tissue factor (1-219) mutants for their ability to support factor Xa-mediated activation of factor VII. The tissue factor residues identified as most important for this interaction (Tyr157, Lys159, Ser163, Gly164, Lys165, Lys166, and Tyr185) were identical to those found to be important for the interaction of substrate factor X with the tissue factor.factor VIIa complex. The residues form a continuous surface-exposed patch with an area of about 500 A(2), which appears to be located outside the tissue factor-factor VII contact zone. In agreement, the two monoclonal antibodies 5G6 and D3H44-F(ab')(2), whose epitopes overlap with this identified region, inhibited the rates of factor VII activation by 86% and 95%, respectively. These antibodies also strongly inhibited the conversion of (125)I-labeled factor VII when cell membrane-expressed, full-length tissue factor (1-263) was employed. Together the results suggest the usage of a common surface region of tissue factor in its dual role-as a cofactor for factor Xa-mediated factor VII activation and as a cofactor for factor VIIa-mediated factor X activation. The finding that factor Xa and factor X may engage in similar, if not identical, molecular interactions with tissue factor further indicates that factor Xa and factor X are similarly oriented toward their respective interaction partners in the ternary catalytic complexes.


Assuntos
Fator VII/metabolismo , Fator Xa/metabolismo , Tromboplastina/metabolismo , Ácido 1-Carboxiglutâmico/metabolismo , Linhagem Celular , Membrana Celular/enzimologia , Membrana Celular/genética , Membrana Celular/metabolismo , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Precursores Enzimáticos/metabolismo , Fator VIIa/metabolismo , Humanos , Radioisótopos do Iodo , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Mapeamento de Peptídeos , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Solubilidade , Ressonância de Plasmônio de Superfície , Tromboplastina/genética
12.
Biochemistry ; 39(25): 7380-7, 2000 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-10858285

RESUMO

The enzymatic activity of coagulation factor VIIa is controlled by its cellular cofactor tissue factor (TF). TF binds factor VIIa with high affinity and, in addition, participates in substrate interaction through its C-terminal fibronectin type III domain. We analyzed surface-exposed residues in the C-terminal TF domain to more fully determine the area on TF important for substrate activation. Soluble TF (sTF) mutants were expressed in E. coli, and their ability to support factor VIIa-dependent substrate activation was measured in the presence of phospholipid vesicles or SW-13 cell membranes. The results showed that factor IX and factor X interacted with the same TF region located proximal to the putative phospholipid surface. According to the degree of activity loss of the sTF mutants, this TF region can be divided into a main region (residues Tyr157, Lys159, Ser163, Gly164, Lys165, Lys166, Tyr185) forming a solvent-exposed patch of 488 A(2) and an extended region which comprises an additional 7-8 residues, including the distally positioned Asn199, Arg200, and Asp204. Some of the identified TF residues, such as Trp158 and those within the loop Lys159-Lys165, are near the factor VIIa gamma-carboxyglutamic acid (Gla) domain, suggesting that the factor VIIa Gla-domain may also participate in substrate interaction. Moreover, the surface identified as important for substrate interaction carries a net positive charge, suggesting that charge interactions may significantly contribute to TF-substrate binding. The calculated surface-exposed area of this substrate interaction region is about 1100 A(2), which is approximately half the size of the TF area that is in contact with factor VIIa. Therefore, a substantial portion of the TF surface (3000 A(2)) is engaged in protein-protein interactions during substrate catalysis.


Assuntos
Fator IX/metabolismo , Fator X/metabolismo , Tromboplastina/metabolismo , Amidas/metabolismo , Anticorpos/imunologia , Ativação Enzimática , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Fosfolipídeos/metabolismo , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Tromboplastina/química , Tromboplastina/genética , Tromboplastina/imunologia
13.
Blood Coagul Fibrinolysis ; 11(1): 27-42, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10691097

RESUMO

The use of libraries of phage-displayed human single-chain antibody fragments (scFv) has become a new, powerful tool in rapidly obtaining therapeutically useful antibodies. Here, we describe the generation of human scFv and F(ab')2 directed against the gamma-carboxyglutamic acid (Gla) domain of coagulation factor IX. A large library of human scFv, displayed either on M13 phage or expressed as soluble proteins, was screened for binding to human Gla-domain peptide (Tyr1-Lys43). Among a panel of scFv that bound to the factor IX-Gla domain, six scFv clones recognized full-length factor IX and exhibited strong inhibitory activity of factor IX in vitro. After reformatting as F(ab')2, the affinity for factor IX of three selected clones was determined: 10C12 Kd = 1.6 nmol/l, 13D1 Kd = 2.9 nmol/l, and 13H6 Kd = 0.46 nmol/l. The antibodies specifically bound to factor IX and not to other coagulation factors, as assessed by enzyme-linked immunosorbent-type and human plasma clotting assays. The complementarity determining region amino acid sequences of clones 10C12 and 13D1 only differed at a single residue, whereas 13H6 showed little homology, suggesting that 13H6 binds to a different epitope within the factor IX-Gla domain. Despite the slightly lower affinity of 10C12 F(ab')2 versus 13H6 F(ab')2, 10C12 was consistently more potent than 13H6 in prolonging the activated partial thromboplastin time (APTT), in inhibiting platelet-mediated plasma clotting, and in inhibiting factor X activation by the intrinsic Xase complex. Finally, 10C12 F(ab')2 also recognized and neutralized factor IX/factor IXa of different species, as demonstrated by the specific APTT prolongation of dog, mouse, baboon and rabbit plasma. In summary, the results validate the usefulness of scFv phage-displayed libraries to rapidly generate fully human antibodies as potential new therapeutics for thrombotic disorders.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/metabolismo , Fator IXa/imunologia , Biblioteca de Peptídeos , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Afinidade de Anticorpos , Reações Antígeno-Anticorpo/efeitos dos fármacos , Sítios de Ligação de Anticorpos , Ligação Competitiva , Coagulação Sanguínea/efeitos dos fármacos , Fatores de Coagulação Sanguínea/metabolismo , Plaquetas/fisiologia , Cálcio/farmacologia , Bovinos , Clonagem Molecular , Endotélio Vascular/citologia , Ensaio de Imunoadsorção Enzimática , Epitopos/metabolismo , Fator IXa/antagonistas & inibidores , Fator IXa/fisiologia , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Fragmentos Fab das Imunoglobulinas/farmacologia , Região Variável de Imunoglobulina/isolamento & purificação , Região Variável de Imunoglobulina/metabolismo , Região Variável de Imunoglobulina/farmacologia , Magnésio/farmacologia , Dados de Sequência Molecular , Alinhamento de Sequência , Especificidade da Espécie
14.
Thromb Haemost ; 84(6): 1072-81, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11154116

RESUMO

Tissue factor (TF), the cellular cofactor for the serine protease factor VIIa (F.VIIa), triggers blood coagulation and is involved in the pathogenesis of various thrombosis-related disorders. Therefore, agents which specifically target tissue factor, such as monoclonal antibodies, may provide promising new antithrombotic therapy. We mapped the epitopes of several anti-TF antibodies using a panel of soluble TF mutants. They bound to three distinct TF regions. The epitope of the 7G11 antibody included Phe50 and overlapped with a TF-F.VIIa light chain contact area. The common epitope of the antibodies 6B4 and HTF1 included residues Tyr94 and Phe76 both of which make critical contacts to the catalytic domain of F.VIIa. The antibodies D3 and 5G6 had a common epitope outside the TF-F.VIIa contact region. It included residues Lys 165, Lys 166, Asn199, Arg200 and Lys201 and thus overlapped with the substrate interaction region of tissue factor. The antibodies 5G6 and D3 were potent anticoagulants when infused to flowing human blood in an ex-vivo thrombosis model. Plasma fibrinopeptide A levels and fibrin deposition were completely inhibited. In contrast, 6B4 was a weak inhibitor in this ex-vivo thrombosis model, and HTF1 displayed no inhibition at all. These disparate activities were also reflected in TF-dependent F.X activation assays performed with human plasma. The potency differences could neither be explained by the determined binding affinities nor by the on-rates of antibodies. Therefore, the results suggest that antibody binding epitope and hence the particular mechanism of inhibition, is the main determinative factor of anticoagulant potency of anti-TF antibodies.


Assuntos
Anticorpos Monoclonais/farmacologia , Epitopos/análise , Tromboplastina/imunologia , Afinidade de Anticorpos , Anticoagulantes/farmacologia , Sítios de Ligação , Coagulação Sanguínea/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo , Cristalografia por Raios X , Epitopos/farmacologia , Fator VIIa/metabolismo , Humanos , Mutação , Tromboplastina/genética , Tromboplastina/metabolismo
15.
Thromb Haemost ; 82(3): 1188-95, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10494786

RESUMO

10C12, a human antibody F(ab')2, which specifically binds to the Gla domain of factor IX, interfered with all known coagulation processes that involve factor IX/IXa. These include the function of the intrinsic Xase complex and the activation of zymogen factor IX by factor XIa and by the tissue factor:factor VIla complex. Furthermore, 10C12 potently inhibited activated partial thromboplastin clotting times (APTT) in plasma of guinea pig and rat, thus enabling in-vivo evaluation. In guinea pigs, a bolus administration of 10C12 (10 microg/kg) prevented cyclic flow variations in damaged carotid arteries without affecting coagulation or bleeding parameters. At a 100-fold higher dose, 10C12 had no effect on normal hemostasis as assessed by the cuticle bleeding time. At this dose, 10C12 was also efficacious in a rat arterial thrombosis model, substantially reducing clot weight and duration of vessel occlusion while prolonging ex-vivo APTT only 1.2-fold. The dose of heparin required to produce comparable antithrombotic effects prolonged the APTT by 12-fold and increased the tail bleeding time (TBT) by 8-fold. In contrast, 10C12 had no effect on TBT. However, rat tails showed a tendency for rebleeding which 10C12 exacerbated. In conclusion, the antithrombotic potency of the 10C12 antibody in two species provides evidence for an important role of F.IX, and its Gla domain in particular, during thrombogenesis under arterial flow conditions. The relative safety at effective doses of this fully human antibody suggests that it may have therapeutic value for treatment of thrombotic disorders.


Assuntos
Ácido 1-Carboxiglutâmico/imunologia , Fator IX/imunologia , Fator IX/metabolismo , Fibrinolíticos/metabolismo , Fibrinolíticos/farmacologia , Fragmentos Fab das Imunoglobulinas/metabolismo , Fragmentos Fab das Imunoglobulinas/farmacologia , Animais , Sítios de Ligação , Coagulação Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Fator IX/antagonistas & inibidores , Cobaias , Hemostasia/efeitos dos fármacos , Heparina/farmacologia , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Trombose/sangue , Trombose/terapia
17.
Thromb Haemost ; 78(3): 1142-9, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9308768

RESUMO

Inhibition of the tissue factor/factor VIIa (TF/F.VIIa) complex attenuates thrombosis in different animal models of arterial thrombosis. However, it remains unclear to what extent the antithrombotic effects are associated with changes in hemostatic functions and how this compares with inhibition of thrombin, an enzyme acting at a later stage in the coagulation cascade. The antithrombotic and the antihemostatic effects of a monoclonal anti-TF antibody (AP-1) were compared in a model of arterial thrombosis to those of a direct thrombin inhibitor (napsagatran) and heparin. In anesthetized rabbits transient arterial thrombi were induced by mechanical damage to the subendothelium of a moderately stenosed carotid artery. Recurrent formation and dislodgement of thrombi resulted in cyclic flow variations (CFVs) which were monitored over 2 hours. Rabbits received intravenously either a placebo (control), a monoclonal anti-rabbit TF antibody (AP-1, 0.05 mg/kg as an i.v. bolus repeated every 15 min, a specific low molecular weight thrombin inhibitor (napsagatran, 3 microg/kg/min) or heparin (3 and 13 microg/kg/min). The effect of the inhibitors on the hemostatic system was studied in a separate set of rabbits by measuring template bleeding times (BT) in the ear arterioles, marginal ear vein and the nail cuticle of the foreleg. AP-1 and napsagatran showed a similar antithrombotic activity (78% and 80% abolition of the CFVs, respectively), whereas either low or high dose heparin was poorly effective (43% and 40% inhibition of CFVs, respectively). At these antithrombotic doses and even at 4-fold higher dosage, AP-1 did not significantly alter the BT, whereas napsagatran and heparin prolonged the ear vessels and cuticle BT in a dose-dependent manner. These results suggest that in contrast to direct thrombin inhibition, the blockade of the TF/F. VIIa function did not result in a concomitant prolongation of the bleeding time. Thus, dissociation of antithrombotic and antihemostatic effects indicates that inhibition of the coagulation system at its initial stage represents a promising approach for the development of new anticoagulants.


Assuntos
Tromboplastina/fisiologia , Trombose/fisiopatologia , Animais , Anticorpos Monoclonais/metabolismo , Anticoagulantes/farmacologia , Antitrombinas/farmacologia , Tempo de Sangramento , Coagulação Sanguínea , Modelos Animais de Doenças , Hemodinâmica , Heparina/farmacologia , Naftalenos/farmacologia , Piperidinas/farmacologia , Coelhos , Trombina/antagonistas & inibidores , Trombina/fisiologia , Tromboplastina/antagonistas & inibidores , Tromboplastina/imunologia
18.
Blood ; 89(4): 1270-8, 1997 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9028950

RESUMO

The adhesion of leukocytes to platelets deposited at the site of vascular injury may represent an important mechanism by which leukocytes contribute to hemostasis and thrombosis. In this study, we examined whether, in comparison with their distribution in circulating blood, certain leukocyte types are enriched at sites of platelet deposition. We used an experimental vascular injury model, in which human fibrillar collagen was exposed to anticoagulated human whole blood flowing through parallel-plate chambers (venous shear rate, 65/s). The platelet-adherent leukocytes were detached by EDTA treatment and analyzed by flow cytometry using cell-type-specific antibodies. The predominant leukocytes found in platelet thrombi were polymorphonuclear leukocytes, accounting for 76% of bound leukocytes (62% in circulating blood), whereas T and B lymphocytes did not significantly accumulate on thrombi, comprising a fraction of less than 5% (32% in circulating blood). Monocytes constituted 16% of platelet thrombus-bound leukocytes, which represents an almost fourfold enrichment as compared with their proportion in circulating blood. Almost identical results were obtained when we analyzed leukocytes adhering to platelet monolayers, which were formed by blocking glycoprotein IIb-IIIa, thus preventing platelet aggregation on top of the collagen-adherent platelets. Furthermore, leukocyte adhesion to platelet monolayers was completely inhibited by an anti-P-selectin antibody (50% inhibitory concentration, 0.3 microg/mL), whereas it reached a plateau at about 70% inhibition on platelet thrombi. This difference could be explained by a possible function of glycoprotein IIb-IIIa in leukocyte immobilization to thrombi or by the high local concentration of P-selectin in the growing thrombi. The results suggest that, because of their known abilities to promote coagulation and thrombolysis, the monocytes and polymorphonuclear leukocytes accumulating on forming platelet thrombi could play an important role in modulating thrombotic and hemostatic processes.


Assuntos
Monócitos/citologia , Neutrófilos/citologia , Adesividade Plaquetária , Trombose/patologia , Adesão Celular , Colágeno/química , Hemorreologia , Humanos , Linfócitos/citologia , Selectina-P/biossíntese , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia
19.
Trends Cardiovasc Med ; 7(8): 316-24, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21235903

RESUMO

The tissue factor:factor VIIa (TF-F.VIIa) complex is considered the physiological initiator of blood coagulation. Besides its role in normal hemostasis, this enzyme complex has been found to play an important role in various thrombotic disorders and thus has become an attractive target for the development of new anticoagulants. Recently, significant progress has been made in regard to structural and molecular aspects of TF-VIIa-initiated coagulation. A rather complete picture on how tissue factor binds to factor VIIa has emerged and is discussed in detail in this review. Also, the combined data of the TF-F.VIIa crystal structure, of naturally occurring F.VII variants, and of mutagenesis studies provide a framework to discuss molecular aspects of the tissue factor-mediated enhancement of F.VIIa catalytic efficiency and the recognition of macromolecular substrates. F.VIIa as a member of the serine protease family has an active site homologous to other coagulation factors. The release of the coordinates of the crystal structures of F.X and F.IX, together with the earlier determined thrombin structure, now allows a detailed comparison of these active centers with respect to the development of specific and potent active site inhibitors. This structural and molecular information about the TF-F.VIIa complex and other coagulation enzymes adds to our understanding of blood coagulation and should further the development of new classes of anticoagulants. (Trends Cardiovasc Med 1997;7:316-324). © 1997, Elsevier Science Inc.

20.
Curr Opin Biotechnol ; 7(4): 386-91, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8768895

RESUMO

Tissue factor (TF), a transmembrane glycoprotein, functions as an essential activator of the serine protease factor VIIa. This enzymatic complex is considered to be the principal initiator of in vivo coagulation. Recent studies emphasize the role of the TF/VIIa complex in a number of pathophysiological processes, such as Gram-negative sepsis, coronary artery disease and neointimal hyperplasia after angioplasty. Monocytes/macrophages are important contributors to some of these diseases and there have been new insights into the biology of TF regulation in monocytes. In the light of its structural similarity to cytokine receptors, there has been frequent speculation that TF has a role in intracellular signaling, a suggestion that is supported by some recent studies that propose a true receptor function for TF.


Assuntos
Coagulação Sanguínea/fisiologia , Fator VIIa/química , Fator VIIa/fisiologia , Tromboplastina/química , Tromboplastina/fisiologia , Animais , Membrana Celular/química , Membrana Celular/fisiologia , Humanos , Modelos Moleculares , Monócitos/química , Monócitos/metabolismo , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA