Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Haemophilia ; 21(6): 761-5, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25930174

RESUMO

INTRODUCTION: Haemophilia A is an X-linked recessive bleeding disorder that primarily affects males. Emerging data support evidence for increased bleeding in female haemophilia A carriers despite factor VIII activity within the normal range. AIM: Data regarding the effect of increased bleeding on health-related quality of life (HR-QOL) in haemophilia A carriers is sparse. We tested the hypothesis that haemophilia A carriers have reduced HR-QOL related to bleeding symptoms. METHODS: We conducted a cross-sectional study at Vanderbilt University. Case subjects were obligate or genetically verified haemophilia A carriers age 18-60 years. Control subjects were mothers of children with cancer who receive care at the Vanderbilt paediatric haematology-oncology clinic. Trained interviewers administered the Rand 36-Item Health Survey 1.0, a validated questionnaire evaluating eight health concepts that may affect HR-QOL, to each study participant. Mann-Whitney U-tests were used to compare median scores for the eight health domains between the case and control groups. RESULT: Forty-two haemophilia A carriers and 36 control subjects were included in analyses. Haemophilia A carriers had significantly lower median scores for the domains of 'Pain' (73.75 vs. 90; P = 0.02) and 'General health' (75 vs. 85; P = 0.01) compared to control subjects. CONCLUSION: Haemophilia A carriers in our study demonstrated significantly lower median scores on the Rand 36-item Health Survey 1.0 in the domains of 'Pain' and 'General Health' compared to women in the control group. Our findings highlight the need for further investigation of the effect of bleeding on HR-QOL in this population.


Assuntos
Saúde , Hemofilia A/genética , Heterozigoto , Qualidade de Vida , Adolescente , Adulto , Estudos Transversais , Feminino , Hemofilia A/complicações , Hemorragia/complicações , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
3.
Cell Mol Bioeng ; 14(2): 161-175, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33868498

RESUMO

INTRODUCTION: Vascular devices such as stents, hemodialyzers, and membrane oxygenators can activate blood coagulation and often require the use of systemic anticoagulants to selectively prevent intravascular thrombotic/embolic events or extracorporeal device failure. Coagulation factor (F)XII of the contact activation system has been shown to play an important role in initiating vascular device surface-initiated thrombus formation. As FXII is dispensable for hemostasis, targeting the contact activation system holds promise as a significantly safer strategy than traditional antithrombotics for preventing vascular device-associated thrombosis. OBJECTIVE: Generate and characterize anti-FXII monoclonal antibodies that inhibit FXII activation or activity. METHODS: Monoclonal antibodies against FXII were generated in FXII-deficient mice and evaluated for their binding and anticoagulant properties in purified and plasma systems, in whole blood flow-based assays, and in an in vivo non-human primate model of vascular device-initiated thrombus formation. RESULTS: A FXII antibody screen identified over 400 candidates, which were evaluated in binding studies and clotting assays. One non-inhibitor and six inhibitor antibodies were selected for characterization in functional assays. The most potent inhibitory antibody, 1B2, was found to prolong clotting times, inhibit fibrin generation on collagen under shear, and inhibit platelet deposition and fibrin formation in an extracorporeal membrane oxygenator deployed in a non-human primate. CONCLUSION: Selective contact activation inhibitors hold potential as useful tools for research applications as well as safe and effective inhibitors of vascular device-related thrombosis.

7.
Science ; 253(5022): 909-12, 1991 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-1652157

RESUMO

Coagulation factor XI is activated in vitro by factor XIIa in the presence of high molecular weight kininogen (HMWK) and a negatively charged surface. Factor XII deficiency is not associated with bleeding, which suggests that another mechanism for factor XI activation exists in vivo. A revised model of coagulation is proposed in which factor XI is activated by thrombin. In the absence of cofactors, thrombin is more effective (kcat/Km = 1.6 x 10(5)) than factor XIIa (1.7 x 10(4)) in activating factor XI. Dextran sulfate enhances activation of factor XI by thrombin 2000-fold; part of this effect is due to autoactivation of factor XI by activated factor XI.


Assuntos
Coagulação Sanguínea , Fator XI/metabolismo , Modelos Biológicos , Compostos Cromogênicos/metabolismo , Sulfato de Dextrana/farmacologia , Fator XI/química , Fator XIIa/farmacologia , Hemostasia/fisiologia , Cininogênios/farmacologia , Substâncias Macromoleculares , Oligopeptídeos/metabolismo , Ácido Pirrolidonocarboxílico/análogos & derivados , Receptores de Superfície Celular/farmacologia , Receptores de Trombina , Proteínas Recombinantes/farmacologia , Trombina/farmacologia
8.
J Thromb Haemost ; 16(10): 2044-2049, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30007049

RESUMO

Essentials Mice lacking factor IX (FIX) or factor XI (FXI) were tested in a saphenous vein bleeding model. FIX-deficient mice displayed a hemostatic defect and FXI-deficient mice were similar to wild type mice. Infusion of FXI or over-expression of FXI in FIX-deficient mice improved hemostasis. FXI may affect the phenotype of FIX-deficiency (hemophilia B). SUMMARY: Background In humans, deficiency of coagulation factor XI may be associated with a bleeding disorder, but, until recently, FXI-deficient mice did not appear to have a hemostatic abnormality. A recent study, however, indicated that FXI-deficient mice show a moderate hemostatic defect in a saphenous vein bleeding (SVB) model. Objectives To study the effect of FXI on bleeding in mice with normal levels of the FXI substrate FIX and in mice lacking FIX (a murine model of hemophilia B). Methods Wild-type mice and mice lacking either FIX (F9- ) or FXI (F11-/- ) were tested in the SVB model. The plasma levels of FXI in F11-/- mice were manipulated by infusion of FXI or its active form FXIa, or by overexpressing FXI by the use of hydrodynamic tail vein injection. Results F9- mice showed a significant defect in the SVB model, whereas F11-/- mice and wild-type mice were indistinguishable. Intravenous infusion of FXI or FXIa into, or overexpression of FXI in, F9- mice improved hemostasis in the SVB model. Overexpression of a FXI variant lacking a FIX-binding site also improved hemostasis in F9- mice. Conclusions Although we were unable to demonstrate a hemostatic defect in F11-/- mice in the SVB model, our results support the premise that supraphysiological levels of FXI improve hemostasis in F9- mice through FIX-independent pathways.


Assuntos
Deficiência do Fator XI/tratamento farmacológico , Fator XI/administração & dosagem , Hemofilia B/tratamento farmacológico , Hemostasia/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Fator IX/genética , Fator IX/metabolismo , Fator XI/genética , Fator XI/metabolismo , Deficiência do Fator XI/sangue , Deficiência do Fator XI/genética , Predisposição Genética para Doença , Hemofilia B/sangue , Hemofilia B/genética , Hemostasia/genética , Infusões Intravenosas , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo
9.
J Thromb Haemost ; 5(6): 1106-12, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17388803

RESUMO

The classic intrinsic pathway of coagulation is triggered by contact activation of the plasma protease factor (F)XII, followed by sequential proteolytic activation of FX1 and FIX. While a key mechanism for initiating coagulation in some clinically useful in vitro assays, the absence of abnormal bleeding associated with congenital FXII deficiency indicates that the intrinsic pathway is not important for normal blood coagulation in vivo. However, recent work with mice lacking FXII or FXI suggest that these proteases make important contributions to formation of pathologic intravascular thrombi. In models of arterial injury, FXII or FXI null mice are protected from formation of platelet rich occlusive thrombi to a degree similar to that seen in FIX deficient mice (a model for the severe bleeding disorder hemophilia B) or to wild type mice treated with high dose heparin. FXII or FXI deficiency does not appear to prevent the initiation of thrombus formation in these models, but instead causes significant thrombus instability that prevents occlusion of the vessel. These findings raise the possibility that a pathway similar or identical to the intrinsic pathway may operate in vivo under some circumstances. Furthermore, the disproportionate importance of FXII and FXI to occlusive thrombus formation compared to normal hemostasis makes these proteases attractive candidates for therapeutic inhibitors to treat or prevent thromboembolic disorders.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/fisiologia , Tromboembolia/sangue , Tromboembolia/tratamento farmacológico , Animais , Anticoagulantes/uso terapêutico , Modelos Animais de Doenças , Fator XI/fisiologia , Fator XII/fisiologia , Humanos , Camundongos , Modelos Cardiovasculares , Tromboplastina/fisiologia
10.
J Thromb Haemost ; 5(4): 781-7, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17229051

RESUMO

Inherited deficiency of the trypsin-like protease factor (F) XI is associated with a mild to moderate bleeding diathesis. In most cases, FXI protein is reduced in plasma, and examples of dysfunctional circulating FXI variants are rare. We characterized the defect in one such variant with a proline to leucine substitution at residue 520. FXI Pro520 corresponds to chymotrypsin Pro161, and is conserved in most members of the chymotrypsin protease family. Recombinant FXI containing this substitution will be referred to as FXI(P161L). k(cat) for cleavage of chromogenic substrates and for activation of the natural FXIa substrate FIX is approximately 3-fold lower for activated FXI(P161L) (FXIa(P161L)) than for wild-type FXIa (FXIa(WT)), consistent with an abnormal protease active site. Inhibition of FXIa(P161L) by diisopropyl fluorophosphate is 2.4-fold slower than for FXIa(WT), suggesting distortion of the protease oxyanion hole. Binding to p-aminobenzamidine, a probe for the integrity of the S1 substrate-binding site, was similar for FXIa(WT) and FXIa(P161L). Rates of carbamylation of Ile16 were also similar for FXIa(WT) and FXIa(P161L), indicating that the critical salt bridge between Ile16 and Asp194 forms normally during protease activation. Cumulatively, the data demonstrate that Pro161 is required for normal active site oxyanion hole conformation in FXIa. Examination of the FXIa crystal structure and modeling studies indicate that Pro161 forms several hydrophobic contacts with adjacent amino acids that stabilize active site conformation. Leucine can be incorporated at position 161 in FXIa, but would not form the extensive stabilizing network of hydrophobic interactions formed by Pro161.


Assuntos
Fator XI/genética , Fator XI/fisiologia , Ácido Aspártico/química , Sítios de Ligação , Coagulação Sanguínea , Catálise , Quimotripsina/química , Humanos , Cinética , Leucina/química , Modelos Moleculares , Prolina/química , Ligação Proteica , Proteínas Recombinantes/química , Serina Endopeptidases/química
11.
Thromb Res ; 156: 134-141, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28644959

RESUMO

BACKGROUND: The plasma protease factor XIa (FXIa) has become a target of interest for therapeutics designed to prevent or treat thrombotic disorders. METHODS: We used a solution-based, directed evolution approach called systematic evolution of ligands by exponential enrichment (SELEX) to isolate RNA aptamers that target the FXIa catalytic domain. RESULTS: Two aptamers, designated 11.16 and 12.7, were identified that bound to previously identified anion binding and serpin bindings sites on the FXIa catalytic domain. The aptamers were non-competitive inhibitors of FXIa cleavage of a tripeptide chromogenic substrate and of FXIa activation of factor IX. In normal human plasma, aptamer 12.7 significantly prolonged the aPTT clotting time. CONCLUSIONS: The results show that novel inhibitors of FXIa can be prepared using SELEX techniques. RNA aptamers can bind to distinct sites on the FXIa catalytic domain and noncompetitively inhibit FXIa activity toward its primary macromolecular substrate factor IX with different levels of potency. Such compounds can be developed for use as therapeutic inhibitors.


Assuntos
Anticoagulantes/metabolismo , Aptâmeros de Nucleotídeos/metabolismo , Fator XIa/metabolismo , Humanos
12.
J Thromb Haemost ; 4(5): 1011-6, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16689752

RESUMO

BACKGROUND: The tissue factor (TF) factor (F) VIIa complex activates coagulation FIX and FX to initiate coagulation, and also cleaves protease activated receptors (PARs) to initiate inflammatory processes in vascular cells. Tissue factor pathway inhibitor (TFPI) is the only specific inhibitor of the TF-FVIIa complex, regulating both its procoagulant and pro-inflammatory properties. Upon heparin infusion during cardiopulmonary bypass (CPB), a heparin releasable pool of endothelial associated TFPI circulates in plasma. Following protamine neutralization of heparin, the plasma TFPI level decreases, but does not return completely to baseline, suggesting that during CPB a fraction of the plasma TFPI becomes heparin-independent. We have investigated the structural and functional properties of plasma TFPI during CPB to further characterize how TFPI is altered during this procedure. METHODS: We enrolled 17 patients undergoing first-time cardiac surgery involving CPB. Plasma samples were obtained at baseline, 5 min and 1 h after start of CPB (receiving heparin), 10 min after protamine administration (off CPB) and 24 h following surgery. Samples were analyzed for full-length and free (non-lipoprotein bound) TFPI antigen by enzyme-linked immunosorbent assay (ELISA) and for TFPI anticoagulant activity using an amidolytic assay. Western blot analysis was used to identify TFPI species of varying molecular weights in three additional patients. Dunnett's test for post hoc comparisons was used for statistical analysis. RESULTS: The ELISA and Western blot data indicated that an increase in full-length TFPI accounted for most of the heparin releasable TFPI. Following heparin neutralization with protamine, the full-length TFPI antigen returned to baseline levels while the free TFPI antigen and the total plasma TFPI activity remained elevated. This was associated with the appearance of a new 38 kDa form of plasma TFPI identified by Western blot analysis. The 38 kDa form of TFPI did not react with an antibody directed against the C-terminal region of TFPI indicating it has undergone proteolysis within this region. All TFPI measurements returned to baseline 24 h following CPB. CONCLUSIONS: During CPB the full-length form of TFPI is the predominant form in plasma because of its prompt release from the endothelial surface following heparin administration. Upon heparin neutralization with protamine, full-length TFPI redistributes back to the endothelial surface. However, a new 38 kDa TFPI fragment is generated during CPB and remains circulating in plasma, indicating that TFPI undergoes proteolytic degradation during CPB. This degradation may result in a decrease in endothelium-associated TFPI immediately post-CPB, and may contribute to the procoagulant and proinflammatory state that often complicates CPB.


Assuntos
Ponte de Artéria Coronária , Lipoproteínas/fisiologia , Adulto , Sequência de Aminoácidos , Anticorpos/imunologia , Western Blotting , Ensaio de Imunoadsorção Enzimática , Antagonistas de Heparina/farmacologia , Humanos , Imunoprecipitação , Lipoproteínas/sangue , Lipoproteínas/química , Lipoproteínas/imunologia , Dados de Sequência Molecular , Protaminas/administração & dosagem
13.
J Thromb Haemost ; 4(9): 1982-8, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16961605

RESUMO

BACKGROUND: Increased plasma levels of coagulation factor (F) XI are a risk factor for venous thrombosis. OBJECTIVE: To further explore the relationship between FXI and venous thrombosis, we evaluated FXI-deficient and wild-type mice in a ferric chloride (FeCl(3))-induced vena cava thrombosis model. METHODS AND RESULTS: Thrombosis was induced by 3-min topical application of filter papers containing increasing concentrations of FeCl(3) and the thrombus was measured at 30 min. In contrast to wild-type mice, FXI-deficient mice failed to form a thrombus with 5% FeCl(3,) and were partially protected against 7.5% and 10% FeCl(3,) respectively. The protective effect was substantially stronger than a high dose of heparin (1,000 units kg(-1), i.v.), clopidogrel (30 mg kg(-1), p.o.) or argatroban (30 mg kg(-1), i.p.). These antithrombotic agents resulted in off-scale bleeding in a tail bleeding time assay, whereas the bleeding time of FXI-deficient mice was unchanged compared to wild-type mice. In addition to its known effect on the coagulation cascade, enhanced clot lysis was demonstrated in FXI-deficient mouse and human plasma compared to those supplemented with FXIa. CONCLUSION: Given the strong antithrombotic efficacy (possibly contributed by strong anticoagulant activity associated with increased fibrinolytic activity) and mild bleeding diathesis associated with FXI deficiency, therapeutic inhibition of FXI may be a reasonable therapeutic strategy to treat or prevent venous thrombosis.


Assuntos
Deficiência do Fator XI/complicações , Compostos Férricos/farmacologia , Veias Cavas/patologia , Trombose Venosa/prevenção & controle , Animais , Cloretos , Modelos Animais de Doenças , Fibrinólise , Fibrinolíticos/farmacologia , Camundongos , Trombose Venosa/induzido quimicamente
14.
Cancer Res ; 50(10): 2895-900, 1990 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-2334893

RESUMO

Exposure of HL-60 promyelocytes to the phorbol ester 12-O-tetradecanoylphorbol-13-acetate increased incorporation of 32P into a Mr approximately 33,000 protein (NP33) found in the nuclear matrices prepared by treating cells with Triton X-100, nucleases, and 2 M NaCl (D. E. Macfarlane, J. Biol. Chem., 261: 6947-6953, 1986). We now report that 12-O-tetradecanoylphorbol-13-acetate causes phosphorylation of NP33 in U937, K562, HEL, Molt-3, and Raji cell lines, all of which are rapidly proliferating cells of hematopoietic origin. 12-O-Tetradecanoylphorbol-13-acetate caused a lesser degree of NP33 phosphorylation in peripheral blood lymphocytes and monocytes and none in granulocytes or platelets. The incorporation of 32P into NP33 was complete in about 10 min, and it was prevented or reversed by staurosporin, indicating that NP33 is continuously phosphorylated and dephosphorylated. NP33 was purified to homogeneity from Triton X-100-washed nuclei or whole cells by extraction with H2SO4, acetone precipitation, and preparative two-dimensional gel electrophoresis. The amino-terminal amino acid sequence of NP33 appears to be the same as that of ribosomal S6 protein. NP33 appears to be S6 protein copurifying with the nuclear matrix.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas Ribossômicas/metabolismo , Alcaloides/farmacologia , Sequência de Aminoácidos , Linhagem Celular , Eletroforese em Gel Bidimensional , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/análise , Fosfoproteínas/análise , Fosforilação , Inibidores de Proteínas Quinases , Proteína S6 Ribossômica , Proteínas Ribossômicas/análise , Estaurosporina , Acetato de Tetradecanoilforbol/farmacologia
15.
Cancer Res ; 49(19): 5329-33, 1989 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-2670202

RESUMO

The human promyelocytic leukemia cell line HL-60 has an amplified number of copies of the protooncogene c-myc. It is induced to differentiate by exposure to the phorbol ester 12-O-tetradecanoyl phorbol-13-acetate (TPA). We have developed a mutant phorbol ester-tolerant (PET) line of HL-60 which undergoes a transient growth arrest but does not differentiate when exposed to TPA (Macfarlane et al., Br. J. Haematol., 68: 291-302, 1988). The defect is not due to a general failure of TPA-induced phosphorylation. In this paper, we show that exposing phorbol ester-sensitive (S) HL-60 cells to TPA caused the disappearance of the c-myc protein antigen (detected on Western blots) in 4 h, whereas TPA had no effect on the c-myc protein content of PET cells. Dimethyl sulfoxide caused the rapid disappearance of the myc antigen in both cells. PET cells had slightly more copies of the c-myc gene detected on Southern blots than S cells. c-myc mRNA was equally unstable in both cells, as determined by Northern blots following actinomycin D. TPA induced the down-regulation of c-myc mRNA in S cells to a greater extent than in PET cells. Dimethyl sulfoxide caused a rapid down-regulation of c-myc mRNA in both cell lines. This shows that PET cells have a defect in the mechanism by which protein kinase C regulates c-myc transcription. Our results provide further evidence that reduction in c-myc expression is necessary for differentiation to occur in HL-60 cells.


Assuntos
Amplificação de Genes , Regulação da Expressão Gênica , Leucemia Promielocítica Aguda/genética , Proteínas Proto-Oncogênicas/genética , Southern Blotting , Diferenciação Celular/efeitos dos fármacos , Dimetil Sulfóxido/farmacologia , Amplificação de Genes/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Promielocítica Aguda/metabolismo , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myc , RNA Mensageiro/análise , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas/metabolismo
16.
J Thromb Haemost ; 14(4): 828-38, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26748875

RESUMO

BACKGROUND: Allosteric inhibition is a promising approach for developing a new group of anticoagulants with potentially reduced bleeding consequences. Recently, we designed sulfated ß-O4 lignin (SbO4L) as an allosteric inhibitor that targets exosite 2 of thrombin to reduce fibrinogen cleavage through allostery and compete with glycoprotein Ibα to reduce platelet activation. OBJECTIVE: To assess: (i) the antithrombotic potential of a novel approach of simultaneous exosite 2-dependent allosteric inhibition of thrombin and competitive inhibition of platelet activation; and (ii) the promise of SbO4L as the first-in-class antithrombotic agent. METHODS: A combination of whole blood thromboelastography, hemostasis analysis, mouse arterial thrombosis models and mouse tail bleeding studies were used to assess antithrombotic potential. RESULTS AND CONCLUSIONS: SbO4L extended the clot initiation time, and reduced maximal clot strength, platelet contractile force, and the clot elastic modulus, suggesting dual anticoagulant and antiplatelet effects. These effects were comparable to those observed with enoxaparin. A dose of 1 mg of SbO4L per mouse prevented occlusion in 100% of arteries, and lower doses resulted in a proportionally reduced response. Likewise, the time to occlusion increased by ~ 70% with a 0.5-mg dose in the mouse Rose Bengal thrombosis model. Finally, tail bleeding studies demonstrated that SbO4L does not increase bleeding propensity. In comparison, a 0.3-mg dose of enoxaparin increased the bleeding time and blood volume loss. Overall, this study highlights the promise of the allosteric inhibition approach, and presents SbO4L as a novel anticoagulant with potentially reduced bleeding side effects.


Assuntos
Anticoagulantes/química , Inibidores da Agregação Plaquetária/química , Complexo Glicoproteico GPIb-IX de Plaquetas/química , Trombina/química , Sítio Alostérico , Animais , Sítios de Ligação , Tempo de Sangramento , Coagulação Sanguínea/efeitos dos fármacos , Testes de Coagulação Sanguínea , Simulação por Computador , Enoxaparina/farmacologia , Fibrinolíticos/química , Hemorragia , Hemostasia , Heparina/uso terapêutico , Humanos , Hidrólise , Lignina/química , Camundongos , Camundongos Endogâmicos C57BL , Ativação Plaquetária , Agregação Plaquetária/efeitos dos fármacos , Plasma Rico em Plaquetas , Ligação Proteica , Risco , Tromboelastografia , Trombina/imunologia , Trombose
17.
J Thromb Haemost ; 3(4): 695-702, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15733058

RESUMO

Factor XI (FXI) and factor IX (FIX) are zymogens of plasma serine proteases required for normal hemostasis. The purpose of this work was to evaluate FXI and FIX as potential therapeutic targets by means of a refined ferric chloride (FeCl(3))-induced arterial injury model in factor-deficient mice. Various concentrations of FeCl(3) were used to establish the arterial thrombosis model in C57BL/6 mice. Carotid artery blood flow was completely blocked within 10 min in C57BL/6 mice by application of 3.5% FeCl(3). In contrast, FXI- and FIX-deficient mice were fully protected from occlusion induced by 5% FeCl(3), and were partially protected against the effect of 7.5% FeCl(3). The protective effect was comparable to very high doses of heparin (1000 units kg(-1)) and substantially more effective than aspirin. While FXI and FIX deficiencies were indistinguishable in the carotid artery injury model, there was a marked difference in a tail-bleeding-time assay. FXI-deficient and wild-type mice have similar bleeding times, while FIX deficiency was associated with severely prolonged bleeding times (>5.8-fold increase, P < 0.01). Given the relatively mild bleeding diathesis associated with FXI deficiency, therapeutic inhibition of FXI may be a reasonable strategy for treating or preventing thrombus formation.


Assuntos
Artérias Carótidas/efeitos dos fármacos , Fator IX/fisiologia , Deficiência do Fator XI/patologia , Fator XI/fisiologia , Compostos Férricos/farmacologia , Hemofilia B/patologia , Animais , Artérias/efeitos dos fármacos , Artérias/lesões , Aspirina/farmacologia , Tempo de Sangramento , Velocidade do Fluxo Sanguíneo , Doenças das Artérias Carótidas/patologia , Cloretos , Relação Dose-Resposta a Droga , Genótipo , Heparina/química , Heparina/farmacologia , Homozigoto , Camundongos , Camundongos Endogâmicos C57BL , Agregação Plaquetária , Fluxo Sanguíneo Regional/efeitos dos fármacos , Trombose/patologia , Trombose/terapia , Fatores de Tempo
18.
Trends Cardiovasc Med ; 10(5): 198-204, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11282295

RESUMO

Blood coagulation factor IX is activated during hemostasis by two distinct mechanisms. Activation through factor VIIa/tissue factor occurs early in the course of fibrin clot formation. Activation by factor XIa appears to be important for maintaining the integrity of the clot over time. In general, coagulation proteases are activated on a phospholipid surface in the presence of a protein cofactor. Until recently, activation of factor IX by factor XIa was thought to be the exception to this rule, as phospholipid has no effect on the reaction and no cofactor had been identified. These curious observations suggest that factor IX is activated by factor XIa in the fluid phase. A large amount of new evidence now indicates that factor IX activation by factor XIa occurs on the surface of activated platelets. The data suggest, however, that this reaction differs significantly from other protease-substrate interactions on the platelet surface. This is likely to be due, in part, to the unusual structure of the factor XI molecule.


Assuntos
Ativação Enzimática , Fator IX/fisiologia , Fator IXa/fisiologia , Sequência de Aminoácidos , Plaquetas/fisiologia , Fator IX/genética , Fator IXa/genética , Humanos , Dados de Sequência Molecular
19.
J Thromb Haemost ; 13(8): 1383-95, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25976012

RESUMO

The most commonly used anticoagulants produce therapeutic antithrombotic effects either by inhibiting thrombin or factor Xa (FXa) or by lowering the plasma levels of the precursors of these key enzymes, prothrombin and FX. These drugs do not distinguish between thrombin generation contributing to thrombosis from thrombin generation required for hemostasis. Thus, anticoagulants increase bleeding risk, and many patients who would benefit from therapy go untreated because of comorbidities that place them at unacceptable risk for hemorrhage. Studies in animals demonstrate that components of the plasma contact activation system contribute to experimentally induced thrombosis, despite playing little or no role in hemostasis. Attention has focused on FXII, the zymogen of a protease (FXIIa) that initiates contact activation when blood is exposed to foreign surfaces, and FXI, the zymogen of the protease FXIa, which links contact activation to the thrombin generation mechanism. In the case of FXI, epidemiologic data indicate this protein contributes to stroke and venous thromboembolism, and perhaps myocardial infarction, in humans. A phase 2 trial showing that reduction of FXI may be more effective than low molecular weight heparin at preventing venous thrombosis during knee replacement surgery provides proof of concept for the premise that an antithrombotic effect can be uncoupled from an anticoagulant effect in humans by targeting components of contact activation. Here, we review data on the role of FXI and FXII in thrombosis and results of preclinical and human trials for therapies targeting these proteins.


Assuntos
Anticoagulantes/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Fator XII/antagonistas & inibidores , Fator XI/antagonistas & inibidores , Fibrinolíticos/uso terapêutico , Trombose Venosa/tratamento farmacológico , Animais , Anticoagulantes/efeitos adversos , Modelos Animais de Doenças , Desenho de Fármacos , Ativação Enzimática , Fator XI/metabolismo , Fator XII/metabolismo , Fibrinolíticos/efeitos adversos , Hemorragia/induzido quimicamente , Humanos , Terapia de Alvo Molecular , Fatores de Risco , Resultado do Tratamento , Trombose Venosa/sangue , Trombose Venosa/diagnóstico
20.
J Thromb Haemost ; 1(11): 2340-7, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14629467

RESUMO

The coagulation protease zymogen factor (F)XI is a disulfide bond-linked homodimer, a configuration that is necessary for protein secretion and function. The non-catalytic portion of the FXI polypeptide contains four repeats called apple domains (A1-A4). It is clear that FXI A4 plays a key role in dimer formation, however, the importance of other apple domains to this process has not been examined. We prepared recombinant FXI molecules in which apple domains were exchanged with those of the structurally homologous monomeric protein prekallikrein (PK). As expected, FXI/PK chimeras containing FXI A4 are dimers, while those with PK A4 are monomers. FXI A4 contains cysteine at position 321 that forms the interchain disulfide bond, while Cys321 in PK is unavailable for interchain bond formation because it is paired with Cys326. FXI/PK chimeras containing PK A4 were modified by changing Cys326 to glycine, leaving Cys321 unpaired (PKA4-Gly326). FXI with a PK A4 domain is a monomer, however, introducing PKA4-Gly326 results in a disulfide bond-linked dimer. This indicates that dimer formation can occur in the absence of FXI A4. In proteins containing PKA4-Gly326, replacing FXI A3 with PK A3 partially interferes with dimer formation, while substitution of A2, or A2 and A3 prevents dimer formation. PKA4-Gly326 cannot induce the native PK molecule to dimerize. The data indicate that FXI A2 and A3 make contributions to dimer formation. As these domains are involved in activities that require dimeric protein, it seems reasonable that they stabilize this conformation.


Assuntos
Fator IX/química , Sítios de Ligação , Cromatografia em Gel , Clonagem Molecular , Dimerização , Dissulfetos , Eletroforese em Gel de Poliacrilamida , Fator IX/genética , Humanos , Pré-Calicreína/química , Pré-Calicreína/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA