Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Thromb Haemost ; 22(1): 225-237, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37813198

RESUMO

BACKGROUND: In plasma, high molecular weight kininogen (HK) is either free or bound to prekallikrein (PK) or factor (F) XI (FXI). During contact activation, HK is thought to anchor PK and FXI to surfaces, facilitating their conversion to the proteases plasma kallikrein and FXIa. Mice lacking HK have normal hemostasis but are resistant to injury-induced arterial thrombosis. OBJECTIVES: To identify amino acids on the HK-D6 domain involved in PK and FXI binding and study the importance of the HK-PK and HK-FXI interactions to coagulation. METHODS: Twenty-four HK variants with alanine replacements spanning residues 542-613 were tested in PK/FXI binding and activated partial thromboplastin time clotting assays. Surface-induced FXI and PK activation in plasma were studied in the presence or absence of HK. Kng1-/- mice lacking HK were supplemented with human or murine HK and tested in an arterial thrombosis model. RESULTS: Overlapping binding sites for PK and FXI were identified in the HK-D6 domain. HK variants with defects only in FXI binding corrected the activated partial thromboplastin time of HK-deficient plasma poorly compared to a variant defective only in PK-binding. In plasma, HK deficiency appeared to have a greater deleterious effect on FXI activation than PK activation. Human HK corrected the defect in arterial thrombus formation in HK-deficient mice poorly due to a specific defect in binding to mouse FXI. CONCLUSION: Clinical observations indicate FXI is required for hemostasis, while HK is not. Yet, the HK-FXI interaction is required for contact activation-induced clotting in vitro and in vivo suggesting an important role in thrombosis and perhaps other FXI-related activities.


Assuntos
Cininogênio de Alto Peso Molecular , Trombose , Animais , Humanos , Camundongos , Cininogênio de Alto Peso Molecular/metabolismo , Fator XI/metabolismo , Pré-Calicreína/metabolismo , Coagulação Sanguínea
2.
J Thromb Haemost ; 21(6): 1567-1579, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36863563

RESUMO

BACKGROUND: During plasma contact activation, factor XII (FXII) binds to surfaces through its heavy chain and undergoes conversion to the protease FXIIa. FXIIa activates prekallikrein and factor XI (FXI). Recently, we showed that the FXII first epidermal growth factor-1 (EGF1) domain is required for normal activity when polyphosphate is used as a surface. OBJECTIVES: The aim of this study was to identify amino acids in the FXII EGF1 domain required for polyphosphate-dependent FXII functions. METHODS: FXII with alanine substitutions for basic residues in the EGF1 domain were expressed in HEK293 fibroblasts. Wild-type FXII (FXII-WT) and FXII containing the EGF1 domain from the related protein Pro-HGFA (FXII-EGF1) were positive and negative controls. Proteins were tested for their capacity to be activated, and to activate prekallikrein and FXI, with or without polyphosphate, and to replace FXII-WT in plasma clotting assays and a mouse thrombosis model. RESULTS: FXII and all FXII variants were activated similarly by kallikrein in the absence of polyphosphate. However, FXII with alanine replacing Lys73, Lys74, and Lys76 (FXII-Ala73,74,76) or Lys76, His78, and Lys81 (FXII-Ala76,78,81) were activated poorly in the presence of polyphosphate. Both have <5% of normal FXII activity in silica-triggered plasma clotting assays and have reduced binding affinity for polyphosphate. Activated FXIIa-Ala73,74,76 displayed profound defects in surface-dependent FXI activation in purified and plasma systems. FXIIa-Ala73,74,76 reconstituted FXII-deficient mice poorly in an arterial thrombosis model. CONCLUSION: FXII Lys73, Lys74, Lys76, and Lys81 form a binding site for polyanionic substances such as polyphosphate that is required for surface-dependent FXII function.


Assuntos
Fator XII , Trombose , Humanos , Animais , Camundongos , Fator XII/metabolismo , Pré-Calicreína/metabolismo , Polifosfatos , Células HEK293 , Fator XI/metabolismo , Fator XIIa/metabolismo
4.
Methods Mol Biol ; 2303: 789-805, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34626423

RESUMO

Evaluating prospective anticoagulant therapies in animal thrombosis and bleeding models are standard pre-clinical approaches. Mice are frequently used for initial evaluations because a variety of models have been developed in this well-characterized species, and mice are relatively inexpensive to maintain. Because mice seem to be resistant to forming "spontaneous" thrombosis, vessel injury is used to induce intravascular clot formation. For the purpose of testing heparin-based drugs, we adapted a well-established model in which thrombus formation in the carotid artery is induced by exposing the vessel to ferric chloride. For studying anticoagulant effects on venous thrombosis, we use a model in which the inferior vena cava is ligated and the size of the resulting clots are measured. The most common adverse effect of anticoagulation therapy is bleeding. We describe a simple tail bleeding time that has been used for many years to study the effects of anticoagulants on hemostasis. We also describe a more reproducible, but more technically challenging, saphenous vein bleeding model that is also used for this purpose.


Assuntos
Anticoagulantes/química , Trombose , Animais , Anticoagulantes/farmacologia , Modelos Animais de Doenças , Hemorragia , Heparitina Sulfato , Camundongos , Estudos Prospectivos , Trombose/tratamento farmacológico
5.
Blood ; 135(8): 558-567, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31800958

RESUMO

Prekallikrein (PK) is the precursor of the trypsin-like plasma protease kallikrein (PKa), which cleaves kininogens to release bradykinin and converts the protease precursor factor XII (FXII) to the enzyme FXIIa. PK and FXII undergo reciprocal conversion to their active forms (PKa and FXIIa) by a process that is accelerated by a variety of biological and artificial surfaces. The surface-mediated process is referred to as contact activation. Previously, we showed that FXII expresses a low level of proteolytic activity (independently of FXIIa) that may initiate reciprocal activation with PK. The current study was undertaken to determine whether PK expresses similar activity. Recombinant PK that cannot be converted to PKa was prepared by replacing Arg371 with alanine at the activation cleavage site (PK-R371A, or single-chain PK). Despite being constrained to the single-chain precursor form, PK-R371A cleaves high-molecular-weight kininogen (HK) to release bradykinin with a catalytic efficiency ∼1500-fold lower than that of kallikrein cleavage of HK. In the presence of a surface, PK-R371A converts FXII to FXIIa with a specific activity ∼4 orders of magnitude lower than for PKa cleavage of FXII. These results support the notion that activity intrinsic to PK and FXII can initiate reciprocal activation of FXII and PK in solution or on a surface. The findings are consistent with the hypothesis that the putative zymogens of many trypsin-like proteases are actually active proteases, explaining their capacity to undergo processes such as autoactivation and to initiate enzyme cascades.


Assuntos
Coagulação Sanguínea , Bradicinina/metabolismo , Pré-Calicreína/metabolismo , Substituição de Aminoácidos , Animais , Fator XII/metabolismo , Células HEK293 , Humanos , Cininogênio de Alto Peso Molecular/metabolismo , Camundongos Endogâmicos C57BL , Pré-Calicreína/química , Pré-Calicreína/genética , Proteólise , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
6.
J Thromb Haemost ; 17(9): 1449-1460, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31125187

RESUMO

BACKGROUND: The homologous plasma proteins prekallikrein and factor XI (FXI) circulate as complexes with high molecular weight kininogen. Although evidence supports an interaction between the prekallikrein-kininogen complexes and vascular endothelium, there is conflicting information regarding FXI binding to endothelium. OBJECTIVE: To study the interaction between FXI and blood vessels in mice. METHODS: C57Bl/6 wild-type or F11-/- mice in which variants of FXI were expressed by hydrodynamic tail vein injection, received intravenous infusions of saline, heparin, polyphosphates, protamine, or enzymes that digest glycosaminoglycans (GAGs). Blood was collected after infusion and plasma was analyzed by western blot for FXI. RESULTS AND CONCLUSIONS: Plasma FXI increased 5- to 10-fold in wild-type mice after infusion of heparin, polyphosphates, protamine, or GAG-digesting enzymes, but not saline. Similar treatments resulted in a much smaller change in plasma FXI levels in rats, and infusions of large boluses of heparin did not change FXI levels appreciably in baboons or humans. The releasable FXI fraction was reconstituted in F11-/- mice by expressing murine FXI, but not human FXI. We identified a cluster of basic residues on the apple 4 domain of mouse FXI that is not present in other species. Replacing the basic residues with alanine prevented the interaction of mouse FXI with blood vessels, whereas introducing the basic residues into human FXI allowed it to bind to blood vessels. Most FXI in mice is noncovalently associated with GAGs on blood vessel endothelium and does not circulate in plasma.


Assuntos
Endotélio Vascular/metabolismo , Fator XI/metabolismo , Glicosaminoglicanos/sangue , Animais , Sítios de Ligação , Trombose das Artérias Carótidas/sangue , Trombose das Artérias Carótidas/induzido quimicamente , Cloretos/toxicidade , Fator XI/química , Deficiência do Fator XI/sangue , Compostos Férricos/toxicidade , Heparina/farmacologia , Humanos , Cininogênios/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Papio , Pré-Calicreína/metabolismo , Ligação Proteica , Conformação Proteica , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Especificidade da Espécie , Eletricidade Estática
7.
Blood ; 133(10): 1152-1163, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30591525

RESUMO

The plasma proteins factor XII (FXII) and prekallikrein (PK) undergo reciprocal activation to the proteases FXIIa and kallikrein by a process that is enhanced by surfaces (contact activation) and regulated by the serpin C1 inhibitor. Kallikrein cleaves high-molecular-weight kininogen (HK), releasing the vasoactive peptide bradykinin. Patients with hereditary angioedema (HAE) experience episodes of soft tissue swelling as a consequence of unregulated kallikrein activity or increased prekallikrein activation. Although most HAE cases are caused by reduced plasma C1-inhibitor activity, HAE has been linked to lysine/arginine substitutions for Thr309 in FXII (FXII-Lys/Arg309). Here, we show that FXII-Lys/Arg309 is susceptible to cleavage after residue 309 by coagulation proteases (thrombin and FXIa), resulting in generation of a truncated form of FXII (δFXII). The catalytic efficiency of δFXII activation by kallikrein is 15-fold greater than for full-length FXII. The enhanced rate of reciprocal activation of PK and δFXII in human plasma and in mice appears to overwhelm the normal inhibitory function of C1 inhibitor, leading to increased HK cleavage. In mice given human FXII-Lys/Arg309, induction of thrombin generation by infusion of tissue factor results in enhanced HK cleavage as a consequence of δFXII formation. The effects of δFXII in vitro and in vivo are reproduced when wild-type FXII is bound by an antibody to the FXII heavy chain (HC; 15H8). The results contribute to our understanding of the predisposition of patients carrying FXII-Lys/Arg309 to angioedema after trauma, and reveal a regulatory function for the FXII HC that normally limits PK activation in plasma.


Assuntos
Fator XII/química , Fator XIa/química , Angioedema Hereditário Tipo III/sangue , Angioedema Hereditário Tipo III/genética , Angioedemas Hereditários , Animais , Arginina/química , Coagulação Sanguínea , Bradicinina/sangue , Catálise , Proteína Inibidora do Complemento C1/química , Fator XIIa/química , Células HEK293 , Humanos , Cininogênios/sangue , Lisina/química , Camundongos , Camundongos Endogâmicos C57BL , Calicreína Plasmática/química , Pré-Calicreína/química , Ligação Proteica , Proteínas Recombinantes/química , Propriedades de Superfície , Trombina/genética
8.
Thromb Res ; 161: 94-105, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29223926

RESUMO

Factor XI (FXI) is the zymogen of a plasma protease, factor XIa (FXIa), that contributes to thrombin generation during blood coagulation by proteolytic activation of several coagulation factors, most notably factor IX (FIX). FXI is a homolog of prekallikrein (PK), a component of the plasma kallikrein-kinin system. While sharing structural and functional features with PK, FXI has undergone adaptive changes that allow it to contribute to blood coagulation. Here we review current understanding of the biology and enzymology of FXI, with an emphasis on structural features of the protein as they relate to protease function.


Assuntos
Fator XI/genética , Fator XI/metabolismo , Humanos , Processamento de Proteína Pós-Traducional
9.
Blood ; 129(11): 1527-1537, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28069606

RESUMO

When blood is exposed to variety of artificial surfaces and biologic substances, the plasma proteins factor XII (FXII) and prekallikrein undergo reciprocal proteolytic conversion to the proteases αFXIIa and α-kallikrein by a process called contact activation. These enzymes contribute to host-defense responses including coagulation, inflammation, and fibrinolysis. The initiating event in contact activation is debated. To test the hypothesis that single-chain FXII expresses activity that could initiate contact activation, we prepared human FXII variants lacking the Arg353 cleavage site required for conversion to αFXIIa (FXII-R353A), or lacking the 3 known cleavage sites at Arg334, Arg343, and Arg353 (FXII-T, for "triple" mutant), and compared their properties to wild-type αFXIIa. In the absence of a surface, FXII-R353A and FXII-T activate prekallikrein and cleave the tripeptide S-2302, demonstrating proteolytic activity. The activity is several orders of magnitude weaker than that of αFXIIa. Polyphosphate, an inducer of contact activation, enhances PK activation by FXII-T, and facilitates FXII-T activation of FXII and FXI. In plasma, FXII-T and FXII-R353A, but not FXII lacking the active site serine residue (FXII-S544A), shortened the clotting time of FXII-deficient plasma and enhanced thrombin generation in a surface-dependent manner. The effect was not as strong as for wild-type FXII. Our results support a model for induction of contact activation in which activity intrinsic to single-chain FXII initiates αFXIIa and α-kallikrein formation on a surface. αFXIIa, with support from α-kallikrein, subsequently accelerates contact activation and is responsible for the full procoagulant activity of FXII.


Assuntos
Coagulação Sanguínea , Fator XII/metabolismo , Proteólise , Domínio Catalítico/genética , Fator XIIa/metabolismo , Humanos , Calicreínas/metabolismo , Propriedades de Superfície
10.
PLoS One ; 11(4): e0152968, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27046148

RESUMO

Sepsis, a systemic inflammatory response to infection, is often accompanied by abnormalities of blood coagulation. Prior work with a mouse model of sepsis induced by cecal ligation and puncture (CLP) suggested that the protease factor XIa contributed to disseminated intravascular coagulation (DIC) and to the cytokine response during sepsis. We investigated the importance of factor XI to cytokine and coagulation responses during the first 24 hours after CLP. Compared to wild type littermates, factor XI-deficient (FXI-/-) mice had a survival advantage after CLP, with smaller increases in plasma levels of TNF-α and IL-10 and delayed IL-1ß and IL-6 responses. Plasma levels of serum amyloid P, an acute phase protein, were increased in wild type mice 24 hours post-CLP, but not in FXI-/- mice, supporting the impression of a reduced inflammatory response in the absence of factor XI. Surprisingly, there was little evidence of DIC in mice of either genotype. Plasma levels of the contact factors factor XII and prekallikrein were reduced in WT mice after CLP, consistent with induction of contact activation. However, factor XII and PK levels were not reduced in FXI-/- animals, indicating factor XI deficiency blunted contact activation. Intravenous infusion of polyphosphate into WT mice also induced changes in factor XII, but had much less effect in FXI deficient mice. In vitro analysis revealed that factor XIa activates factor XII, and that this reaction is enhanced by polyanions such polyphosphate and nucleic acids. These data suggest that factor XI deficiency confers a survival advantage in the CLP sepsis model by altering the cytokine response to infection and blunting activation of the contact (kallikrein-kinin) system. The findings support the hypothesis that factor XI functions as a bidirectional interface between contact activation and thrombin generation, allowing the two processes to influence each other.


Assuntos
Coinfecção/metabolismo , Citocinas/metabolismo , Deficiência do Fator XI/metabolismo , Peptídeo Hidrolases/metabolismo , Sepse/metabolismo , Animais , Coinfecção/enzimologia , Ativação Enzimática , Camundongos , Sepse/enzimologia
11.
Thromb Res ; 140: 118-124, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26950760

RESUMO

Studies with animal models implicate the plasma proteases factor XIIa (FXIIa) and α-kallikrein in arterial and venous thrombosis. As congenital deficiencies of factor XII (FXII) or prekallikrein (PK), the zymogens of FXIIa and α-kallikrein respectively, do not cause bleeding disorders, inhibition of these enzymes may have therapeutic benefit without compromising hemostasis. The relative contributions of FXIIa and α-kallikrein to thrombosis in animal models are not clear. We compared mice lacking FXII or PK to wild type mice in established models of arterial thrombosis. Wild type mice developed carotid artery occlusion when the vessel was exposed to a 3.5% solution of ferric chloride (FeCl3). FXII-deficient mice were resistant to occlusion at 5% FeCl3 and partially resistant at 10% FeCl3. PK-deficient mice were resistant at 3.5% FeCl3 and partially resistant at 5% FeCl3. Mice lacking high molecular weight kininogen, a cofactor for PK activation and activity, were also partially resistant to thrombosis at 5% FeCl3. Induction of carotid artery thrombosis with Rose Bengal was delayed in FXII-deficient mice compared to wild type or PK-deficient animals. In human plasma supplemented with silica, DNA or collagen to induce contact activation, an antibody to the FXIIa active site was more effective at preventing thrombin generation than an antibody to the α-kallikrein active site. Similarly, the FXIIa antibody was more effective at reducing fibrin formation in human blood flowing through collagen coated-tubes. The findings suggest that inhibitors of FXIIa will have more potent anti-thrombotic effects than inhibitors of α-kallikrein.


Assuntos
Transtornos da Coagulação Sanguínea/complicações , Deficiência do Fator XII/complicações , Fator XII/genética , Pré-Calicreína/deficiência , Pré-Calicreína/genética , Trombose/etiologia , Trombose/genética , Animais , Coagulação Sanguínea/efeitos dos fármacos , Transtornos da Coagulação Sanguínea/genética , Fator XII/antagonistas & inibidores , Deficiência do Fator XII/genética , Deleção de Genes , Humanos , Calicreínas/antagonistas & inibidores , Calicreínas/genética , Camundongos , Camundongos Endogâmicos C57BL , Pré-Calicreína/antagonistas & inibidores , Fatores de Proteção , Trombose/prevenção & controle
12.
Blood ; 125(9): 1488-96, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25587039

RESUMO

Activation of coagulation factor XI (FXI) may play a role in hemostasis. The primary substrate of activated FXI (FXIa) is FIX, leading to FX activation (FXa) and thrombin generation. However, recent studies suggest the hemostatic role of FXI may not be restricted to the activation of FIX. We explored whether FXI could interact with and inhibit the activity of tissue factor pathway inhibitor (TFPI). TFPI is an essential reversible inhibitor of activated factor X (FXa) and also inhibits the FVIIa-TF complex. We found that FXIa neutralized both endothelium- and platelet-derived TFPI by cleaving the protein between the Kunitz (K) 1 and K2 domains (Lys86/Thr87) and at the active sites of the K2 (Arg107/Gly108) and K3 (Arg199/Ala200) domains. Addition of FXIa to plasma was able to reverse the ability of TFPI to prolong TF-initiated clotting times in FXI- or FIX-deficient plasma, as well as FXa-initiated clotting times in FX-deficient plasma. Treatment of cultured endothelial cells with FXIa increased the generation of FXa and promoted TF-dependent fibrin formation in recalcified plasma. Together, these results suggest that the hemostatic role of FXIa may be attributed not only to activation of FIX but also to promoting the extrinsic pathway of thrombin generation through inactivation of TFPI.


Assuntos
Coagulação Sanguínea/fisiologia , Plaquetas/metabolismo , Fator IX/metabolismo , Fator XIa/metabolismo , Fator Xa/metabolismo , Fibrina/metabolismo , Lipoproteínas/metabolismo , Plaquetas/citologia , Western Blotting , Células Cultivadas , Citometria de Fluxo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Lipoproteínas/genética , Mutação/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
13.
Methods Mol Biol ; 1229: 483-96, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25325974

RESUMO

Evaluating anticoagulants in animal thrombosis models is a standard component of preclinical drug testing. Mice are frequently used for these initial evaluations because a variety of thrombosis models have been developed and are well characterized in this species, and the animals are relatively inexpensive to maintain. Because mice have a natural resistance to forming intravascular thrombi, vessel injury is required to induce intravascular clot formation. Several methods have been established for inducing arterial or venous thrombosis in mice. For the purpose of testing heparin-based drugs, we adapted a well-established model in which thrombus formation in the carotid artery is induced by exposing the vessel to ferric chloride. For studying anticoagulant effects on venous thrombosis, we use a model in which the inferior vena cava is ligated and the size of the resulting clots is measured. The most common adverse effect of anticoagulation therapy is bleeding. The effect of heparin-based anticoagulants can be tested in mice in a simple tail bleeding assay.


Assuntos
Anticoagulantes/uso terapêutico , Heparitina Sulfato/uso terapêutico , Animais , Anticoagulantes/farmacologia , Cloretos , Modelos Animais de Doenças , Compostos Férricos , Hemorragia/tratamento farmacológico , Heparitina Sulfato/farmacologia , Camundongos Endogâmicos C57BL , Cauda , Trombose/tratamento farmacológico , Veia Cava Inferior/efeitos dos fármacos , Veia Cava Inferior/patologia
14.
Blood ; 123(11): 1739-46, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24408325

RESUMO

The plasma zymogens factor XII (fXII) and factor XI (fXI) contribute to thrombosis in a variety of mouse models. These proteins serve a limited role in hemostasis, suggesting that antithrombotic therapies targeting them may be associated with low bleeding risks. Although there is substantial epidemiologic evidence supporting a role for fXI in human thrombosis, the situation is not as clear for fXII. We generated monoclonal antibodies (9A2 and 15H8) against the human fXII heavy chain that interfere with fXII conversion to the protease factor XIIa (fXIIa). The anti-fXII antibodies were tested in models in which anti-fXI antibodies are known to have antithrombotic effects. Both anti-fXII antibodies reduced fibrin formation in human blood perfused through collagen-coated tubes. fXII-deficient mice are resistant to ferric chloride-induced arterial thrombosis, and this resistance can be reversed by infusion of human fXII. 9A2 partially blocks, and 15H8 completely blocks, the prothrombotic effect of fXII in this model. 15H8 prolonged the activated partial thromboplastin time of baboon and human plasmas. 15H8 reduced fibrin formation in collagen-coated vascular grafts inserted into arteriovenous shunts in baboons, and reduced fibrin and platelet accumulation downstream of the graft. These findings support a role for fXII in thrombus formation in primates.


Assuntos
Modelos Animais de Doenças , Deficiência do Fator XII/complicações , Fator XII/antagonistas & inibidores , Fator XII/fisiologia , Trombina/metabolismo , Trombose/prevenção & controle , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Coagulação Sanguínea , Fator XI/metabolismo , Fator XIIa/metabolismo , Fibrina/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Papio , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Tromboplastina/metabolismo , Trombose/etiologia , Trombose/metabolismo
15.
Blood ; 121(19): 3962-9, 2013 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-23515926

RESUMO

Factor XI (fXI) is a homodimeric zymogen that is converted to a protease with 1 (1/2-fXIa) or 2 (fXIa) active subunits by factor XIIa (fXIIa) or thrombin. It has been proposed that the dimeric structure is required for normal fXI activation. Consistent with this premise, fXI monomers do not reconstitute fXI-deficient mice in a fXIIa-dependent thrombosis model. FXI activation by fXIIa or thrombin is a slow reaction that can be accelerated by polyanions. Phosphate polymers released from platelets (poly-P) can enhance fXI activation by thrombin and promote fXI autoactivation. Poly-P increased initial rates of fXI activation 30- and 3000-fold for fXIIa and thrombin, respectively. FXI monomers were activated more slowly than dimers by fXIIa in the presence of poly-P. However, this defect was not observed when thrombin was the activating protease, nor during fXI autoactivation. The data suggest that fXIIa and thrombin activate fXI by different mechanisms. FXIIa may activate fXI through a trans-activation mechanism in which the protease binds to 1 subunit of the dimer, while activating the other subunit. For activation by thrombin, or during autoactivation, the data support a cis-activation mechanism in which the activating protease binds to and activates the same fXI subunit.


Assuntos
Fator XI/química , Fator XI/metabolismo , Fator XIa/metabolismo , Animais , Trombose das Artérias Carótidas/genética , Trombose das Artérias Carótidas/metabolismo , Fator XI/genética , Deficiência do Fator XI/genética , Deficiência do Fator XI/metabolismo , Fator XIIa/química , Fator XIIa/metabolismo , Fator XIa/química , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína
17.
Blood ; 116(19): 3981-9, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-20634381

RESUMO

Mice lacking factor XII (fXII) or factor XI (fXI) are resistant to experimentally-induced thrombosis, suggesting fXIIa activation of fXI contributes to thrombus formation in vivo. It is not clear whether this reaction has relevance for thrombosis in pri mates. In 2 carotid artery injury models (FeCl(3) and Rose Bengal/laser), fXII-deficient mice are more resistant to thrombosis than fXI- or factor IX (fIX)-deficient mice, raising the possibility that fXII and fXI function in distinct pathways. Antibody 14E11 binds fXI from a variety of mammals and interferes with fXI activation by fXIIa in vitro. In mice, 14E11 prevented arterial occlusion induced by FeCl(3) to a similar degree to total fXI deficiency. 14E11 also had a modest beneficial effect in a tissue factor-induced pulmonary embolism model, indicating fXI and fXII contribute to thrombus formation even when factor VIIa/tissue factor initiates thrombosis. In baboons, 14E11 reduced platelet-rich thrombus growth in collagen-coated grafts inserted into an arteriovenous shunt. These data support the hypothesis that fXIIa-mediated fXI activation contributes to thrombus formation in rodents and primates. Since fXII deficiency does not impair hemostasis, targeted inhibition of fXI activation by fXIIa may be a useful antithrombotic strategy associated with a low risk of bleeding complications.


Assuntos
Fator XIIa/fisiologia , Fator XI/fisiologia , Trombose/sangue , Trombose/etiologia , Animais , Anticorpos Monoclonais/farmacologia , Anticoagulantes/farmacologia , Trombose das Artérias Carótidas/sangue , Trombose das Artérias Carótidas/etiologia , Gatos , Modelos Animais de Doenças , Cães , Fator XI/antagonistas & inibidores , Deficiência do Fator XI/sangue , Deficiência do Fator XI/genética , Deficiência do Fator XI/fisiopatologia , Deficiência do Fator XII/sangue , Deficiência do Fator XII/genética , Deficiência do Fator XII/fisiopatologia , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Papio anubis , Tempo de Tromboplastina Parcial , Embolia Pulmonar/sangue , Embolia Pulmonar/etiologia , Coelhos , Especificidade da Espécie
19.
J Infect Dis ; 198(2): 271-4, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18491973

RESUMO

Anticoagulation is a rational approach to the treatment of sepsis-associated consumptive coagulopathy, but its application is limited because of the risk of excessive bleeding. Factor XI (FXI) contributes substantially to pathological blood coagulation (thrombosis), whereas it contributes only modestly to normal hemostasis. We found that FXI-deficient mice have reduced coagulopathy and increased survival relative to FXI-expressing wild-type mice during cecal ligation and puncture-induced acute peritonitis/sepsis. This finding suggests that FXI contributes to coagulopathy and/or inflammation during sepsis and that pharmacologic inhibition of FXI activity may alter the course and outcome of some infections.


Assuntos
Deficiência do Fator XI/complicações , Doenças Peritoneais/microbiologia , Sepse/complicações , Animais , Coagulação Sanguínea , Ceco/microbiologia , Fator XI/fisiologia , Inflamação/complicações , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritonite/complicações , Peritonite/mortalidade , Punções , Valores de Referência , Sepse/mortalidade , Sobreviventes
20.
Blood ; 106(9): 3055-7, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15985536

RESUMO

Plasminogen (Plg)-deficient mice experience wasting and have decreased longevity due to disseminated fibrin deposition. We generated mice with combined deficiencies of Plg and coagulation factor IX (fIX) or XI (fXI) to determine the effects on the Plg null phenotype. Mice lacking Plg and fIX (Plg(-/-)/fIX-/-) have lower mortality at age 6 months than Plg(-/-)/fIX+/+ mice (15% and 67%, respectively) and less severe wasting, consistent with the importance of fIX in fibrin formation. In contrast, combined Plg and fXI deficiency (Plg(-/-)/fXI-/-) reduces life span (more than 90% mortality at 6 months) and is associated with leukocyte infiltration of the lungs and pulmonary fibrosis. These abnormalities are not seen in Plg-/- or Plg(-/-)/fIX-/- animals. Activated fXI is thought to function primarily as a fIX activator; however, our observation suggests that fXI may have functions in regulation of inflammation or tissue repair distinct from its role in coagulation.


Assuntos
Fator IX/metabolismo , Fator XI/metabolismo , Peptídeo Hidrolases/deficiência , Plasminogênio/deficiência , Plasminogênio/metabolismo , Animais , Fator IX/genética , Fator XI/genética , Inflamação , Longevidade , Pulmão/anormalidades , Pulmão/metabolismo , Camundongos , Camundongos Knockout , Peptídeo Hidrolases/metabolismo , Plasminogênio/genética , Fibrose Pulmonar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...