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.
Expert Opin Emerg Drugs ; 28(1): 43-53, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36927160

RESUMO

INTRODUCTION: Whilst the introduction of direct oral anticoagulants (DOACs) has improved the prevention of thromboembolic events, there is still a need for safer anticoagulants. This is particularly so, for specific populations of patients, such as those with an increased bleeding risk or those with severely reduced kidney function. People with Factor XI (FXI) deficiency are at reduced risk of thromboembolic events, without an increased risk of spontaneous bleeding. FXI inhibition, therefore, presents the ideal target for novel anticoagulants. AREAS COVERED: In this review, we provide an overview of the currently available anticoagulants and the emerging FXIa inhibitors in clinical trials. The need for availability of novel anticoagulants and the potential issues that will hinder the development and marketing of factor XIa inhibitors is also discussed. EXPERT OPINION: Evidence suggests that FXI inhibition presents a promising drug target for novel anticoagulation therapies. The FXIa inhibitors in development have advantages over DOACs with lower renal clearance and long half-lives. Overall, FXI inhibition presents a promising target, it is likely that the clinical use of FXIa inhibitors is on the horizon.


Assuntos
Fator XIa , Tromboembolia , Humanos , Fator XIa/fisiologia , Coagulação Sanguínea , Anticoagulantes/efeitos adversos , Tromboembolia/prevenção & controle , Hemorragia/induzido quimicamente
2.
Thromb Haemost ; 120(6): 883-993, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32375196

RESUMO

Cardiovascular disease, including stroke, myocardial infarction, and venous thromboembolism, is one of the leading causes of morbidity and mortality worldwide. Excessive coagulation may cause vascular occlusion in arteries and veins eventually leading to thrombotic diseases. Studies in recent years suggest that coagulation factors are involved in these pathological mechanisms. Factors XIa (FXIa), XIIa (FXIIa), and plasma kallikrein (PKa) of the contact system of coagulation appear to contribute to thrombosis while playing a limited role in hemostasis. Contact activation is initiated upon autoactivation of FXII on negatively charged surfaces. FXIIa activates plasma prekallikrein (PK) to PKa, which in turn activates FXII and initiates the kallikrein-kinin pathway. FXI is also activated by FXIIa, leading to activation of FIX and finally to thrombin formation, which in turn activates FXI in an amplification loop. Animal studies have shown that arterial and venous thrombosis can be reduced by the inhibition of FXI(a) or PKa. Furthermore, data from human studies suggest that these enzymes may be valuable targets to reduce thrombosis risk. In this review, we discuss the structure and function of FXI(a) and PK(a), their involvement in the development of venous and arterial thrombosis in animal models and human studies, and current therapeutic strategies.


Assuntos
Arteriopatias Oclusivas/sangue , Fator XIa/fisiologia , Calicreína Plasmática/fisiologia , Trombose/sangue , Animais , Coagulação Sanguínea/fisiologia , Transtornos da Coagulação Sanguínea/sangue , Fatores de Coagulação Sanguínea/fisiologia , Modelos Animais de Doenças , Ativação Enzimática , Deficiência do Fator XI/sangue , Fator XIa/química , Fator XIa/imunologia , Inibidores do Fator Xa/uso terapêutico , Humanos , Camundongos , Camundongos Knockout , Pré-Calicreína/deficiência , Pré-Calicreína/metabolismo , Processamento de Proteína Pós-Traducional , Especificidade da Espécie , Trombofilia/tratamento farmacológico , Trombose Venosa/sangue
3.
Arterioscler Thromb Vasc Biol ; 39(7): 1390-1401, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31242030

RESUMO

Objective- Activation of coagulation FXI (factor XI) by FXIIa (activated factor XII) is a prothrombotic process. The endothelium is known to play an antithrombotic role by limiting thrombin generation and platelet activation. It is unknown whether the antithrombotic role of the endothelium includes sequestration of FXIa (activated factor XI) activity. This study aims to determine the role of endothelial cells (ECs) in the regulation of the intrinsic pathway of coagulation. Approach and Results- Using a chromogenic assay, we observed that human umbilical veins ECs selectively blocked FXIa yet supported kallikrein and FXIIa activity. Western blotting and mass spectrometry analyses revealed that FXIa formed a complex with endothelial PAI-1 (plasminogen activator inhibitor-1). Blocking endothelial PAI-1 increased the cleavage of a chromogenic substrate by FXIa and the capacity of FXIa to promote fibrin formation in plasma. Western blot and immunofluorescence analyses showed that FXIa-PAI-1 complexes were either released into the media or trafficked to the early and late endosomes and lysosomes of ECs. When baboons were challenged with Staphylococcus aureus to induce a prothrombotic phenotype, an increase in circulating FXIa-PAI-1 complex levels was detected by ELISA within 2 to 8 hours postchallenge. Conclusions- PAI-1 forms a complex with FXIa on ECs, blocking its activity and inducing the clearance and degradation of FXIa. Circulating FXIa-PAI-1 complexes were detected in a baboon model of S. aureus sepsis. Although ECs support kallikrein and FXIIa activity, inhibition of FXIa by ECs may promote the clearance of intravascular FXIa. Visual Overview- An online visual overview is available for this article.


Assuntos
Coagulação Sanguínea , Células Endoteliais/fisiologia , Fator XIa/fisiologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Animais , Fator XIa/antagonistas & inibidores , Fator XIa/química , Humanos , Papio ursinus , Inibidor 1 de Ativador de Plasminogênio/química
4.
Arterioscler Thromb Vasc Biol ; 39(4): 799-809, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30700130

RESUMO

Objective- Factor XI (FXI) contributes to thrombotic disease while playing a limited role in normal hemostasis. We generated a unique, humanized anti-FXI antibody, AB023, which blocks factor XIIa-mediated FXI activation without inhibiting FXI activation by thrombin or the procoagulant function of FXIa. We sought to confirm the antithrombotic activity of AB023 in a baboon thrombosis model and to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics in healthy adult subjects. Approach and Results- In a primate model of acute vascular graft thrombosis, AB023 reduced platelet and fibrin accumulation within the grafts by >75%. To evaluate the safety of AB023, we performed a first-in-human study in healthy adult volunteers without any serious adverse events. Overall, 10 of 21 (48%) subjects experienced 20 treatment-emergent adverse events, with 7 of 16 (44%) subjects following active treatment and 3 of 5 (60%) subjects following placebo. AB023 did not increase bleeding or prothrombin times. Anticoagulation was verified by a saturable ≈2-fold prolongation of the partial thromboplastin time for over 1 month after the highest dose. Conclusions- AB023, which inhibits contact activation-initiated blood coagulation in vitro and experimental thrombus formation in primates, produced a dose-dependent duration of limited anticoagulation without drug-related adverse effects in a phase 1 trial. When put in context with earlier observations suggesting that FXI contributes to venous thromboembolism and cardiovascular disease, although contributing minimally to hemostasis, our data further justify clinical evaluation of AB023 in conditions where contact-initiated FXI activation is suspected to have a pathogenic role. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT03097341.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticoagulantes/uso terapêutico , Fator XI/antagonistas & inibidores , Fator XIa/fisiologia , Fibrinolíticos/uso terapêutico , Adulto , Animais , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacologia , Anticoagulantes/efeitos adversos , Anticoagulantes/imunologia , Anticoagulantes/farmacologia , Área Sob a Curva , Relação Dose-Resposta a Droga , Método Duplo-Cego , Fator XI/imunologia , Fator XIIa/fisiologia , Fibrinolíticos/efeitos adversos , Fibrinolíticos/imunologia , Fibrinolíticos/farmacologia , Oclusão de Enxerto Vascular/tratamento farmacológico , Humanos , Papio , Tempo de Tromboplastina Parcial
5.
Int J Hematol ; 105(3): 235-243, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27848184

RESUMO

The plasma contact system sits atop the intrinsic coagulation cascade and plasma kallikrein-kinin pathway, and in vivo its activation contributes, respectively, to coagulation and inflammation mainly via two downstream pathways. This system has been widely investigated, its activation mechanisms by negatively charged surfaces and the interactions within its components, factor XII, prekallikrein and high molecular weight kininogen are well understood at the biochemical level. However, as most of the activators that have been discovered by in vitro experiments are exogenous, the physiological activators and roles of the contact system have remained unclear and controversial. In the last two decades, several physiological activators have been identified, and a better understanding of its roles and its connection with other signaling pathways has been obtained from in vivo studies. In this article, we present an overview of the contact pathway with a focus on the activation mechanisms, natural stimuli, possible physiological roles, potential risks of its excessive activation, remaining questions and future prospects.


Assuntos
Coagulação Sanguínea/fisiologia , Animais , Fator XIa/fisiologia , Humanos , Plasma/metabolismo , Transdução de Sinais/fisiologia
6.
J Thromb Haemost ; 11(12): 2118-27, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24152424

RESUMO

BACKGROUND: Factor XIa is traditionally assigned a role in FIX activation during coagulation. However, recent evidence suggests this protease may have additional plasma substrates. OBJECTIVE: To determine whether FXIa promotes thrombin generation and coagulation in plasma in the absence of FIX, and to determine whether FXI-deficiency produces an antithrombotic effect in mice independently of FIX. METHODS: FXIa, FXIa variants and anti-FXIa antibodies were tested for their effects on plasma coagulation and thrombin generation in the absence of FIX, and for their effects on the activation of purified coagulation factors. Mice with combined FIX and FXI deficiency were compared with mice lacking either FIX or FXI in an arterial thrombosis model. RESULTS: In FIX-deficient plasma, FXIa induced thrombin generation, and anti-FXIa antibodies prolonged clotting times. This process involved FXIa-mediated conversion of FX and FV to their active forms. Activation of FV by FXIa required the A3 domain on the FXIa heavy chain, whereas activation of FX did not. FX activation by FXIa, unlike FIX activation, was not a calcium-dependent process. Mice lacking both FIX and FXI were more resistant to ferric chloride-induced carotid artery occlusion than FXI-deficient or FIX-deficient mice. CONCLUSION: In addition to its predominant role as an activator of FIX, FXIa may contribute to coagulation by activating FX and FV. As the latter reactions do not require calcium, they may make important contributions to in vitro clotting triggered by contact activation. The reactions may be relevant to FXIa's roles in hemostasis and in promoting thrombosis.


Assuntos
Coagulação Sanguínea/fisiologia , Fator IX/fisiologia , Fator XIa/fisiologia , Animais , Eletroforese em Gel de Poliacrilamida , Fator IX/imunologia , Fator XIa/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteólise
7.
J Biol Chem ; 286(36): 31904-14, 2011 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-21778227

RESUMO

To select residues in coagulation factor XIa (FXIa) potentially important for substrate and inhibitor interactions, we examined the crystal structure of the complex between the catalytic domain of FXIa and the Kunitz protease inhibitor (KPI) domain of a physiologically relevant FXIa inhibitor, protease nexin 2 (PN2). Six FXIa catalytic domain residues (Glu(98), Tyr(143), Ile(151), Arg(3704), Lys(192), and Tyr(5901)) were subjected to mutational analysis to investigate the molecular interactions between FXIa and the small synthetic substrate (S-2366), the macromolecular substrate (factor IX (FIX)) and inhibitor PN2KPI. Analysis of all six Ala mutants demonstrated normal K(m) values for S-2366 hydrolysis, indicating normal substrate binding compared with plasma FXIa; however, all except E98A and K192A had impaired values of k(cat) for S-2366 hydrolysis. All six Ala mutants displayed deficient k(cat) values for FIX hydrolysis, and all were inhibited by PN2KPI with normal values of K(i) except for K192A, and Y5901A, which displayed increased values of K(i). The integrity of the S1 binding site residue, Asp(189), utilizing p-aminobenzamidine, was intact for all FXIa mutants. Thus, whereas all six residues are essential for catalysis of the macromolecular substrate (FIX), only four (Tyr(143), Ile(151), Arg(3704), and Tyr(5901)) are important for S-2366 hydrolysis; Glu(98) and Lys(192) are essential for FIX but not S-2366 hydrolysis; and Lys(192) and Tyr(5901) are required for both inhibitor and macromolecular substrate interactions.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Domínio Catalítico , Fator XIa/antagonistas & inibidores , Fator XIa/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Aprotinina , Catálise , Humanos , Cinética , Inibidores de Proteases , Estrutura Terciária de Proteína
8.
J Thromb Haemost ; 8(7): 1532-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20456758

RESUMO

SUMMARY BACKGROUND: In the extrinsic pathway, the essential procofactors factor (F) V and FVIII are activated to FVa and FVIIIa by thrombin. In the contact pathway and its clinical diagnostic test, the activated partial thromboplastin time (APTT) assay, the sources of procofactor activation are unknown. In the APTT assay, FXII is activated on a negatively charged surface and proceeds to activate FXI, which activates FIX upon the addition of Ca(2+). FIXa feeds thrombin generation through activation of FX. FIXa is an extremely poor catalyst in the absence of its FVIIIa cofactor, which, in the intrinsic FXase complex, increases FXa generation by approximately 10(7). One potential APTT procofactor activator in this setting is FXIa. OBJECTIVE: To test the hypothesis that FXIa can activate FVIII and FV. METHODS: Recombinant FVIII and plasma FV were treated with FXIa, and the activities and integrities of each procofactor were measured using commercial clotting assays and sodium dodecylsulfate polyacrylamide gel electrophoresis (SDS-PAGE). RESULTS: Kinetic analyses of FXIa-catalyzed activation and inactivation of FV and FVIII are reported, and the the timing and sites of cleavage are defined. CONCLUSIONS: FXIa activates both procofactors at plasma protein concentrations, and computational modeling suggests that procofactor activation during the preincubation phase of the APTT assay is critical to the performance of the assay. As the APTT assay is the primary tool for the diagnosis and management of hemophilias A and B, as well as in the determination of FVIII inhibitors, these findings have potential implications in the clinical setting.


Assuntos
Fator VIII/metabolismo , Fator V/metabolismo , Fator XIa/fisiologia , Sítios de Ligação , Testes de Coagulação Sanguínea/métodos , Testes de Coagulação Sanguínea/normas , Catálise , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Humanos , Cinética , Tempo de Tromboplastina Parcial/métodos , Tempo de Tromboplastina Parcial/normas
9.
Arterioscler Thromb Vasc Biol ; 30(3): 388-92, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20139363

RESUMO

The dose-limiting issue with available anticoagulant therapies is bleeding. Is there an approach that could provide antithrombotic protection with reduced bleeding? One hypothesis is that targeting proteases upstream from the common pathway provides a reduction in thrombin sufficient to impede occlusive thrombosis yet allows enough thrombin generation to support hemostasis. The impairment of intrinsic coagulation by selective inhibition of factor XI (FXI) leaves the extrinsic and common pathways of coagulation intact, making FXI a drug target. This concept is supported by the observation that human deficiency in FXI results in a mild bleeding disorder compared with other coagulation factor deficiencies, and that elevated levels of FXI are a risk factor for thromboembolic disease. Moreover, FXI knockout mice have reduced thrombosis with little effect on hemostasis. The results from genetic models have been supported by studies using neutralizing antibodies, peptide inhibitors, and small-molecule inhibitors. These agents impede thrombosis without affecting bleeding time in a variety of experimental animals, including primates. Together, these data strongly support FXIa inhibition as a viable method to increase the ratio of benefit to risk in an antithrombotic drug.


Assuntos
Anticoagulantes/uso terapêutico , Fator XIa/antagonistas & inibidores , Trombose Venosa/tratamento farmacológico , Animais , Anticoagulantes/efeitos adversos , Coagulação Sanguínea/fisiologia , Modelos Animais de Doenças , Fator XIa/genética , Fator XIa/fisiologia , Hemorragia/etiologia , Hemorragia/fisiopatologia , Hemostasia/fisiologia , Humanos , Camundongos , Camundongos Knockout , Medição de Risco , Trombose Venosa/prevenção & controle
10.
Semin Thromb Hemost ; 35(4): 416-25, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19598070

RESUMO

Severe factor XI (FXI) deficiency is an injury-related bleeding disorder common in Ashkenazi Jews and rare worldwide. In the past two decades, more than 180 mutations in the FXI gene have been reported in patients with FXI deficiency, five of which show a founder effect (Cys38Arg, Gln88Stop, Cys128Stop, Glu117stop, and Phe283Leu, the last two largely prevalent among Ashkenazi Jews). Inhibitors to FXI after exposure to plasma, FXI concentrates, or Rh immunoglobulin were described in patients with mutations resulting in null alleles. Treatment with low-dose recombinant activated factor VII in these patients appears promising. Survival advantages to patients with severe FXI have been recently reported. Herein, we present new observations related to clinic presentation, genotype-phenotype correlation, and treatment problems in patients with FXI deficiency.


Assuntos
Deficiência do Fator XI/genética , Fator XI/genética , Antifibrinolíticos/uso terapêutico , Inibidores dos Fatores de Coagulação Sanguínea , Deficiência do Fator XI/complicações , Deficiência do Fator XI/diagnóstico , Fator XIa/fisiologia , Feminino , Efeito Fundador , Genótipo , Hemorragia/genética , Hemorragia/prevenção & controle , Humanos , Recém-Nascido , Judeus/genética , Masculino , Menorragia/terapia , Mutação , Fenótipo , Hemorragia Pós-Parto/terapia , Gravidez , Diagnóstico Pré-Natal , Cuidados Pré-Operatórios , Trombose/etiologia
11.
Life Sci ; 85(5-6): 220-5, 2009 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-19500599

RESUMO

AIMS: Several activated coagulation factors have been reported to enhance fibrinolysis by inactivating plasminogen activator inhibitor type 1 (PAI-1), a serine protease inhibitor. We analyzed the interaction between PAI-1 and the three serine proteases generated during contact activation of plasma, activated factor XII (FXIIa), FXIa, and kallikrein, and evaluated their effects on fibrinolysis in-vitro. MAIN METHODS: Effects of kaolin on euglobulin clot lysis time (ECLT) and behavior of PAI-1 in factor-depleted plasma were analyzed. KEY FINDINGS: The ECLT of pooled plasma obtained from normal volunteers (designated as 100%) was shortened to 62.1+/-3.1% by Ca(2+) (5 mM) and 29.9+/-3.1% by kaolin. Activated protein C reversed the ECLT shortened by Ca(2+)-supplementation (86.3+/-17.4%), but did not affect the ECLT shortened by kaolin (31.4+/-2.1%). Thus, in contrary to Ca(2+)-supplementation, kaolin appeared to shorten the ECLT by a mechanism independent of thrombin generation. In three kinds of contact factor-depleted plasma, kaolin did not shorten ECLT only in FXII-depleted plasma. PAI-1 was cleaved to its inactive form in the Ca(2+) as well as the kaolin-supplemented euglobulin fraction in normal plasma, the latter of which, however, was not observed in FXII-depleted plasma. Similarly, a high molecular weight complex between FXIIa and PAI-1, as well as a cleaved form of PAI-1, was observed in kaolin-supplemented normal plasma, but neither was found in kaolin-supplemented FXII-depleted plasma. SIGNIFICANCE: PAI-1 inactivation by FXIIa appears to be a mechanism by which contact phase coagulation factors enhance fibrinolysis independently of thrombin generation.


Assuntos
Fator XIIa/fisiologia , Fibrinólise/fisiologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inativadores de Plasminogênio/farmacologia , Inibidores de Serina Proteinase/metabolismo , Testes de Coagulação Sanguínea , Coagulantes/imunologia , Coagulantes/metabolismo , Fator XIa/fisiologia , Humanos , Calicreínas/fisiologia , Caulim/farmacologia , Inativadores de Plasminogênio/imunologia , Soroglobulinas/efeitos dos fármacos , Trombina/efeitos dos fármacos , Trombina/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/imunologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
12.
J Biol Chem ; 283(11): 6696-705, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18192270

RESUMO

Factor XI is the zymogen of a dimeric plasma protease, factor XIa, with two active sites. In solution, and during contact activation in plasma, conversion of factor XI to factor XIa proceeds through an intermediate with one active site (1/2-FXIa). Factor XIa and 1/2-FXIa activate the substrate factor IX, with similar kinetic parameters in purified and plasma systems. During hemostasis, factor IX is activated by factors XIa or VIIa, by cleavage of the peptide bonds after Arg145 and Arg180. Factor VIIa cleaves these bonds sequentially, with accumulation of factor IX alpha, an intermediate cleaved after Arg145. Factor XIa also cleaves factor IX preferentially after Arg145, but little intermediate is detected. It has been postulated that the two factor XIa active sites cleave both factor IX peptide bonds prior to releasing factor IX abeta. To test this, we examined cleavage of factor IX by four single active site factor XIa proteases. Little intermediate formation was detected with 1/2-FXIa, factor XIa with one inhibited active site, or a recombinant factor XIa monomer. However, factor IX alpha accumulated during activation by the factor XIa catalytic domain, demonstrating the importance of the factor XIa heavy chain. Fluorescence titration of active site-labeled factor XIa revealed a binding stoichiometry of 1.9 +/- 0.4 mol of factor IX/mol of factor XIa (Kd = 70 +/- 40 nm). The results indicate that two forms of activated factor XI are generated during coagulation, and that each half of a factor XIa dimer behaves as an independent enzyme with respect to factor IX.


Assuntos
Fator IX/metabolismo , Fator XIa/química , Fator XIa/fisiologia , Arginina/química , Sítios de Ligação , Bioquímica/métodos , Catálise , Domínio Catalítico , Dimerização , Hemostasia , Humanos , Hidrólise , Cinética , Modelos Biológicos , Peptídeos/química , Proteínas Recombinantes/química
13.
Expert Rev Hematol ; 1(1): 87-98, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19714257

RESUMO

Factor IX is a key component of the plasma system that forms a fibrin clot at a site of vascular injury. Activation of factor IX by factor XIa is required in certain situations to prevent bleeding from premature clot degradation. Factor XIa is a coagulation protease comprised of two identical subunits. The biochemical and physiologic implications of this unusual structural feature are being actively investigated. Congenital factor XI deficiency causes a mild-to-moderate bleeding disorder, with hemorrhage typically involving the oral/nasal cavities and the urinary tract. Current treatment recommendations take this tissue-specific bleeding pattern into account and target factor replacement to certain types of procedures and clinical situations. Results from animal models and human population studies indicate that factor XI contributes to thromboembolic disease. This protease may therefore be a legitimate therapeutic target.


Assuntos
Fator IX/fisiologia , Fator XIa/metabolismo , Animais , Modelos Animais de Doenças , Fator IX/química , Fator IX/metabolismo , Deficiência do Fator XI/metabolismo , Deficiência do Fator XI/patologia , Deficiência do Fator XI/terapia , Fator XIa/química , Fator XIa/fisiologia , Humanos , Trombose/patologia
15.
J Thromb Haemost ; 4(7): 1496-501, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16839345

RESUMO

BACKGROUND: Thrombus formation through the activation of tissue factor (TF) and factor (F) XI is a critical event in the onset of cardiovascular disease. TF expressed in atherosclerotic plaques and circulating blood is an important determinant of thrombogenicity that contributes to fibrin-rich thrombus formation after plaque disruption. However, the contribution of FXI to thrombus formation on disrupted plaques remains unclear. METHODS: A mouse monoclonal antibody against FXI and activated FXI (FXIa) (XI-5108) was generated by immunization with activated human FXI. Prothrombin time (PT), activated partial thromboplastin time (APTT), bleeding time, and ex vivo platelet aggregation in rabbits were measured before and after an intravenous bolus injection of XI-5108. We investigated the role of FXI upon arterial thrombus growth in the rabbit iliac artery in the presence of repeated balloon injury. RESULTS: The XI-5108 antibody reacted to the light chain of human and rabbit FXI/FXIa, and inhibited FXIa-initiated FXa and FXIa generation. Fibrin-rich thrombi developed on the injured neointima that was obviously immunopositive for glycoprotein IIb-IIIa, fibrin, TF, and FXI. Intravenous administration of XI-5108 (3.0 mg kg(-1)) remarkably reduced thrombus growth, and the APTT was significantly prolonged. However, PT, bleeding time and platelet aggregation were not affected. CONCLUSIONS: These results indicate that plasma FXI plays a potent role in thrombus growth on the injured neointima. Inhibition of plasma FXI activity might help to reduce thrombus growth on ruptured plaques without prolonging bleeding time.


Assuntos
Fator XI/fisiologia , Fator XIa/fisiologia , Artéria Ilíaca/patologia , Trombose/etiologia , Túnica Íntima/patologia , Angioplastia com Balão/efeitos adversos , Animais , Testes de Coagulação Sanguínea , Humanos , Coelhos , Trombose/prevenção & controle , Túnica Íntima/lesões
16.
Biochemistry ; 44(30): 10416-22, 2005 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-16042419

RESUMO

Factor XI, unlike other coagulation proteins, is a homodimer of two identical subunits linked by a single disulfide bond formed by Cys321. The present study was undertaken to understand the physiological significance of the dimeric nature of factor XI. We have expressed a mutant FXI/G326C in which the Gly326 residue of factor XI has been mutated to Cys326, reasoning that Cys321 would form an intrachain disulfide bond with Cys326 as in prekallikrein, a plasma protein that exists as a monomer even with 58% amino acid sequence identity and a domain structure very similar to factor XI. No free thiol could be detected in the expressed protein, and it migrated as a monomer on nonreduced SDS-PAGE. In physiological buffer, however, the protein was found to exist in a state of monomer-dimer equilibrium as assessed by gel-filtration chromatography and ultracentrifugation studies (K(d) approximately 36 nM). Functional studies revealed that FXI/G326C was indistinguishable from plasma factor XI in a plasma-clotting assay and in a factor IX activation assay both in the presence and absence of activated platelets even at concentrations at which less than 5% of the mutant exists as dimers. We conclude that, for optimal function in the presence of activated platelets, a preformed dimer of factor XI is not required.


Assuntos
Fator IX/metabolismo , Fator XIa/química , Fator XIa/fisiologia , Substituição de Aminoácidos/genética , Linhagem Celular , Cisteína/genética , Sulfato de Dextrana/farmacologia , Dimerização , Fator IX/química , Fator XIIa/fisiologia , Fator XIa/genética , Glicina/genética , Humanos , Hidrólise , Oligopeptídeos/metabolismo , Tempo de Tromboplastina Parcial/métodos , Ativação Plaquetária/genética , Ácido Pirrolidonocarboxílico/análogos & derivados , Ácido Pirrolidonocarboxílico/metabolismo , Especificidade por Substrato/genética , Trombina/fisiologia
17.
Arch Pathol Lab Med ; 126(11): 1382-6, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12421145

RESUMO

OBJECTIVES: To review the literature for conditions, diseases, and disorders that affect activity of the contact factors, and further to review the literature for evidence that less than normal activity of any of the contact factors may be associated with thrombophilia. DATA SOURCES: MEDLINE search for English-language articles published from 1988 to 2001 and pertinent references contained therein, as well as search of references in recent relevant articles and reviews. STUDY SELECTION: Relevant clinical and laboratory information was extracted from selected articles. Meta-analysis was not feasible because of heterogeneity of reports. DATA EXTRACTION AND SYNTHESIS: Evidence for association of altered levels of the contact factors and thrombophilia was sought. A wide variety of disorders is associated with decreased activity of the contact factors; chief among these disorders are liver disease, hepatic immaturity of newborns, the antiphospholipid syndrome, and, for factor XII, being of Asian descent. These disorders are more common than homozygous deficiency. The few series and case reports of thrombophilic events in patients homozygous for deficiency of contact factors are not persuasive enough to support causality. The apparent association between levels consistent with heterozygosity (40%-60% of normal) of any of the contact factors (but especially factor XII) in persons with antiphospholipid antibodies appears to be due to falsely decreased in vitro activity levels of these factors, which are normal on antigenic testing. The apparent association with thrombosis is better explained by the antiphospholipid syndrome than by the modest reduction of the levels of contact factors. CONCLUSIONS: Presently, it is not recommended to measure activity of contact factors during routine evaluation of patients who have suffered venous or arterial thromboembolism or acute coronary syndromes.


Assuntos
Fator XIa/fisiologia , Trombofilia/diagnóstico , Coagulação Sanguínea/fisiologia , Testes de Coagulação Sanguínea , Fator XII/fisiologia , Deficiência do Fator XII/diagnóstico , Deficiência do Fator XII/etiologia , Deficiência do Fator XII/fisiopatologia , Humanos , Cininogênio de Alto Peso Molecular/deficiência , Cininogênio de Alto Peso Molecular/fisiologia , Guias de Prática Clínica como Assunto , Pré-Calicreína/deficiência , Pré-Calicreína/fisiologia , Trombofilia/complicações , Trombofilia/fisiopatologia
18.
Blood ; 97(10): 3117-22, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11342438

RESUMO

Human coagulation factor XI (FXI) is a plasma serine protease composed of 2 identical 80-kd polypeptides connected by a disulfide bond. This dimeric structure is unique among blood coagulation enzymes. The hypothesis was tested that dimeric conformation is required for normal FXI function by generating a monomeric version of FXI (FXI/PKA4) and comparing it to wild-type FXI in assays requiring factor IX activation by activated FXI (FXIa). FXI/PKA4 was made by replacing the FXI A4 domain with the A4 domain from prekallikrein (PK). A dimeric version of FXI/PKA4 (FXI/PKA4-Gly326) was prepared as a control. Activated FXI/PKA4 and FXI/PKA4-Gly326 activate factor IX with kinetic parameters similar to those of FXIa. In kaolin-triggered plasma clotting assays containing purified phospholipid, FXI/PKA4 and FXI/PKA4-Gly326 have coagulant activity similar to FXI. The surface of activated platelets is likely to be a physiologic site for reactions involving FXI/FXIa. In competition binding assays FXI/PKA4, FXI/PKA4-Gly326, and FXI have similar affinities for activated platelets (K(i) = 12-16 nM). In clotting assays in which phospholipid is replaced by activated platelets, the dimeric proteins FXI and FXI/PKA4-Gly326 promote coagulation similarly; however, monomeric FXI/PKA4 has greatly reduced activity. Western immunoblot analysis confirmed that activated monomeric FXI/PKA4 activates factor IX poorly in the presence of activated platelets. These findings demonstrate the importance of the dimeric state to FXI activity and suggest a novel model for factor IX activation in which FXIa binds to activated platelets by one chain of the dimer, while binding to factor IX through the other.


Assuntos
Plaquetas/química , Dimerização , Fator IX/fisiologia , Fator XIa/química , Fator XIa/fisiologia , Coagulação Sanguínea , Plaquetas/fisiologia , Western Blotting , Membrana Celular/química , Ativação Enzimática , Fator IXa/fisiologia , Fator XI/fisiologia , Fator XIa/genética , Humanos , Cinética , Ativação Plaquetária , Pré-Calicreína/química , Pré-Calicreína/genética , Conformação Proteica , Proteínas Recombinantes de Fusão
19.
J Biol Chem ; 275(48): 37340-6, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10973954

RESUMO

In the present studies we have made the novel observation that protease nexin 1 (PN1), a member of the serine protease inhibitor (SERPIN) superfamily, is a potent inhibitor of the blood coagulation Factor XIa (FXIa). The inhibitory complexes formed between PN1 and FXIa are stable when subjected to reducing agents, SDS, and boiling, a characteristic of the acyl linkage formed between SERPINs and their cognate proteases. Using a sensitive fluorescence-quenched peptide substrate, the K(assoc) of PN1 for FXIa was determined to be 7.9 x 10(4) m(-)(1) s(-)(1) in the absence of heparin. In the presence of heparin, this rate was accelerated to 1.7 x 10(6), M(-)(1) s(-)(1), making PN1 a far better inhibitor of FXIa than C1 inhibitor, which is the only other SERPIN known to significantly inhibit FXIa. FXIa-PN1 complexes are shown to be internalized and degraded by human fibroblasts, most likely via the low density lipoprotein receptor-related protein (LRP), since degradation was strongly inhibited by the LRP agonist, receptor-associated protein. Since FXIa proteolytically modifies the amyloid precursor protein, this observation may suggest an accessory role for PN1 in the pathobiogenesis of Alzheimer's disease.


Assuntos
Proteínas de Transporte/farmacologia , Proteínas Inativadoras do Complemento 1/farmacologia , Fator XIa/fisiologia , Heparina/farmacologia , Serpinas/fisiologia , Precursor de Proteína beta-Amiloide , Linhagem Celular , Proteína Inibidora do Complemento C1 , Fator XIa/antagonistas & inibidores , Humanos , Nexinas de Proteases , Receptores de Superfície Celular , Inibidores de Serina Proteinase/farmacologia , Serpina E2
20.
Thromb Haemost ; 79(2): 296-301, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9493579

RESUMO

We have made use of a novel flow reactor to study the initiation and propagation of the ex vivo blood coagulation processes at artificial surfaces. The flow reactor consisted of a primary glass or polymer capillary that is connected to a secondary glass capillary, which inner wall was coated with a phospholipid bilayer of 25 mol% dioleoylphosphatidylserine/75 mol% dioleoylphosphatidylcholine (DOPS/DOPC). Citrated platelet free plasma and a CaCl2 solution were delivered by syringe pumps and mixed just before the entrance of the flow reactor. The outflowing plasma was assayed for factor XIa, factor IXa, factor Xa and thrombin activity. Perfusion of recalcified plasma through a bare glass capillary resulted in a transient generation of fluid phase factor XIa. In contrast, factor IXa production increased slowly to attain a stable steady-state level. We established that surface-bound factor XIa was responsible for a continuous production of factor IXa. Factor IXa-induced generation of factor Xa and thrombin was only observed when contact activated plasma was subsequently perfused through a DOPS/DOPC-coated capillary, showing that propagation of the factor IXa trigger requires a procoagulant, phosphatidylserine-containing, phospholipid membrane. The negatively charged inner surface of a heparin-coated polyurethane capillary, generated like the glass capillary significant amounts of factor XIa and factor IXa when perfused with recalcified plasma. No differences were found between unfractionated heparin and heparin devoid of anticoagulant activity. Thus, it is concluded that contact activation and factor IXa generation in flowing plasma is not inhibited by immobilised anticoagulant active heparin. Consequently, factor IXa-dependent thrombin generation at a downstream located phospholipid membrane was similar, regardless the specific anticoagulant activity of immobilised heparin.


Assuntos
Coagulação Sanguínea , Hemorreologia/instrumentação , Fator IXa/fisiologia , Fator XIa/fisiologia , Fator Xa/fisiologia , Hemorreologia/métodos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...