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










Base de dados
Intervalo de ano de publicação
1.
Clin Pharmacol Drug Dev ; 13(3): 265-280, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38224232

RESUMO

Inhibition of activated factor XI reduces thrombogenesis while maintaining physiological hemostasis, with the expectation of reduced bleeding risk compared with standard of care in the clinical setting. Asundexian (BAY 2433334), an activated factor XI inhibitor, is in clinical development for the prevention of thromboembolic events. The effect of asundexian and its plasma metabolite M10 on cardiac repolarization and potential interactions with the hNav1.5 sodium, hCav1.2 calcium, and human ether-à-go-go-related gene (hERG) potassium channels was investigated in vitro. Additionally, asundexian effects on cardiac parameters and electrocardiogram were examined in telemetered beagle dogs. A randomized, placebo-controlled, 4-way crossover, thorough QT study in healthy adults evaluated the influence of 50 and 150 mg of asundexian on the corrected QT interval, including 400 mg of moxifloxacin as positive control. Across all studies, asundexian and M10 were not associated with any effects on cardiac repolarization. The largest in vitro effects of asundexian (approximately 20% inhibition) were seen for hCav1.2 and hERG. Throughout the thorough QT study, the upper limits of the one-sided 95% confidence interval of placebo-corrected mean changes from baseline in Fridericia corrected QT for 50 and 150 mg of asundexian were below Δ = 10 milliseconds. Asundexian demonstrated favorable safety and tolerability profiles.


Assuntos
Fator XIa , Fluoroquinolonas , Adulto , Humanos , Animais , Cães , Fluoroquinolonas/farmacologia , Fator XIa/farmacologia , Coração , Moxifloxacina/farmacologia , Eletrocardiografia
2.
Blood ; 143(15): 1465-1475, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38142404

RESUMO

ABSTRACT: Direct oral anticoagulants (DOACs) that inhibit the coagulation proteases thrombin or factor Xa (FXa) have replaced warfarin and other vitamin K antagonists (VKAs) for most indications requiring long-term anticoagulation. In many clinical situations, DOACs are as effective as VKAs, cause less bleeding, and do not require laboratory monitoring. However, because DOACs target proteases that are required for hemostasis, their use increases the risk of serious bleeding. Concerns over therapy-related bleeding undoubtedly contribute to undertreatment of many patients who would benefit from anticoagulation therapy. There is considerable interest in the plasma zymogen factor XI (FXI) and its protease form factor XIa (FXIa) as drug targets for treating and preventing thrombosis. Laboratory and epidemiologic studies support the conclusion that FXI contributes to venous and arterial thrombosis. Based on 70 years of clinical observations of patients lacking FXI, it is anticipated that drugs targeting this protein will cause less severe bleeding than warfarin or DOACs. In phase 2 studies, drugs that inhibit FXI or FXIa prevent venous thromboembolism after total knee arthroplasty as well as, or better than, low molecular weight heparin. Patients with heart disease on FXI or FXIa inhibitors experienced less bleeding than patients taking DOACs. Based on these early results, phase 3 trials have been initiated that compare drugs targeting FXI and FXIa to standard treatments or placebo. Here, we review the contributions of FXI to normal and abnormal coagulation and discuss results from preclinical, nonclinical, and clinical studies of FXI and FXIa inhibitors.


Assuntos
Fator XI , Trombose , Humanos , Fator XIa/farmacologia , Varfarina/uso terapêutico , Varfarina/farmacologia , Trombose/tratamento farmacológico , Trombose/etiologia , Trombose/prevenção & controle , Coagulação Sanguínea , Anticoagulantes/efeitos adversos , Hemorragia/induzido quimicamente , Fibrinolíticos/uso terapêutico
3.
Bioorg Chem ; 142: 106951, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37924755

RESUMO

Thrombotic diseases, such as myocardial infarction, stroke, and deep vein thrombosis, severely threaten human health, and anticoagulation is an effective way to prevent such illnesses. However, most anticoagulant drugs in the clinic have different bleeding risks. Previous studies have shown that coagulation factor XI is an ideal target for safe anticoagulant drug development. Here, we designed the FXIa inhibitory peptide DX-88mut by replacing Loop1 (DGPCRAAHPR) and Loop2 (IYGGC) in DX-88, which is a clinical drug targeting PKa for the treatment of hereditary angioedema, using Loop1 (TGPCRAMISR) and Loop2 (FYGGC) in the FXIa inhibitory peptide PN2KPI, respectively. DX-88mut selectively inhibited FXIa against a panel of serine proteases with an IC50 value of 14.840 ± 0.453 nM, dose-dependently prolonged APTT in mouse, rat and human plasma, and potently inhibited FeCl3-induced carotid artery thrombosis in mice at a dose of 1 µmol/kg. Additionally, DX-88mut did not show a significant bleeding risk at a dose of 5 µmol/kg. Taken together, these results show that DX-88mut is a potential candidate for the development of a novel antithrombotic agent.


Assuntos
Fator XIa , Trombose , Humanos , Ratos , Camundongos , Animais , Fator XIa/metabolismo , Fator XIa/farmacologia , Coagulação Sanguínea , Anticoagulantes/farmacologia , Trombose/tratamento farmacológico , Peptídeos/farmacologia
4.
Blood ; 136(6): 740-748, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32369559

RESUMO

The bispecific antibody emicizumab is increasingly used for hemophilia A treatment. However, its specificity for human factors IX and X (FIX and FX) has limited its in vivo functional analysis to primate models of acquired hemophilia. Here, we describe a novel mouse model that allows emicizumab function to be examined. Briefly, FVIII-deficient mice received IV emicizumab 24 hours before tail-clip bleeding was performed. A second infusion with human FIX and FX, administered 5 minutes before bleeding, generated consistent levels of emicizumab (0.7-19 mg/dL for 0.5-10 mg/kg doses) and of both FIX and FX (85 and 101 U/dL, respectively, after dosing at 100 U/kg). Plasma from these mice display FVIII-like activity in assays (diluted activated partial thromboplastin time and thrombin generation), similar to human samples containing emicizumab. Emicizumab doses of 1.5 mg/kg and higher significantly reduced blood loss in a tail-clip-bleeding model using FVIII-deficient mice. However, reduction was incomplete compared with mice treated with human FVIII concentrate, and no difference in efficacy between doses was observed. From this model, we deducted FVIII-like activity from emicizumab that corresponded to a dose of 4.5 U of FVIII per kilogram (ie, 9.0 U/dL). Interestingly, combined with a low FVIII dose (5 U/kg), emicizumab provided enough additive activity to allow complete bleeding arrest. This model could be useful for further in vivo analysis of emicizumab.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Fator IX/administração & dosagem , Fator X/administração & dosagem , Hemofilia A/tratamento farmacológico , Hemorragia/tratamento farmacológico , Modelos Animais , Animais , Anticorpos Biespecíficos/administração & dosagem , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/imunologia , Quimioterapia Combinada , Fator IX/análise , Fator IX/imunologia , Fator VIII/administração & dosagem , Fator VIII/análise , Fator VIII/uso terapêutico , Fator X/análise , Fator X/imunologia , Fator XIa/farmacologia , Feminino , Hemofilia A/sangue , Hemofilia A/complicações , Hemofilia A/imunologia , Hemorragia/etiologia , Infusões Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tempo de Tromboplastina Parcial , Cauda/lesões , Trombina/biossíntese
6.
BMC Biotechnol ; 18(1): 21, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29621998

RESUMO

BACKGROUND: Hirudin is a potent thrombin inhibitor but its antithrombotic properties are offset by bleeding side-effects. Because hirudin's N-terminus must engage thrombin's active site for effective inhibition, fusing a cleavable peptide at this site may improve hirudin's risk/benefit ratio as a therapeutic agent. Previously we engineered a plasmin cleavage site (C) between human serum albumin (HSA) and hirudin variant 3 (HV3) in fusion protein HSACHV3. Because coagulation factor XI (FXI) is more involved in thrombosis than hemostasis, we hypothesized that making HV3 activity FXIa-dependent would also improve HV3's potential therapeutic profile. We combined albumin fusion for half-life extension of hirudin with positioning of an FXIa cleavage site N-terminal to HV3, and assessed in vitro and in vivo properties of this novel protein. RESULTS: FXIa cleavage site EPR was employed. Fusion protein EPR-HV3HSA but not HSAEPR-HV3 was activated by FXIa in vitro. FVIIa, FXa, FXIIa, or plasmin failed to activate EPR-HV3HSA. FXIa-cleavable EPR-HV3HSA reduced the time to occlusion of ferric chloride-treated murine arteries and reduced fibrin deposition in murine endotoxemia; noncleavable mycHV3HSA was without effect. EPR-HV3HSA elicited less blood loss than constitutively active HV3HSA in murine liver laceration or tail transection but extended bleeding time to the same extent. EPR-HV3HSA was partially activated in citrated human or murine plasma to a greater extent than HSACHV3. CONCLUSIONS: Releasing the N-terminal block to HV3 activity using FXIa was an effective way to limit hirudin's bleeding side-effects, but plasma instability of the exposed EPR blocking peptide rendered it less useful than previously described plasmin-activatable HSACHV3.


Assuntos
Fator XIa/farmacologia , Hemorragia/prevenção & controle , Hirudinas/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Trombose/tratamento farmacológico , Albuminas/biossíntese , Albuminas/farmacologia , Animais , Fator XIa/biossíntese , Hirudinas/biossíntese , Camundongos , Modelos Animais
7.
PLoS One ; 11(2): e0148255, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26840952

RESUMO

INTRODUCTION: Hemophilia B is an inherited X chromosome-linked disorder characterized by impaired blood clotting owing to the absence of functional coagulation factor IX. Due to the relatively short half-life of factor IX, patients with hemophilia B require frequent factor IX infusions to maintain prophylaxis. We have developed a recombinant factor IX (rFIX) fused to the Fc region of IgG (rFIXFc) with an extended half-life in animals and humans. MATERIALS AND METHODS: Procoagulant properties of rFIXFc and rFIX (BENEFIX®) were compared to determine the effect of the Fc region on rFIXFc hemostatic function. Specifically, we assessed rFIXFc activation, intermolecular interactions within the Xase complex, inactivation by antithrombin III (AT) and thrombin generation potential compared with rFIX. We also assessed the acute and prophylactic efficacy profiles of rFIXFc and rFIX in vivo in hemophilia B mouse bleeding models. RESULTS AND CONCLUSIONS: The activation by factor XIa or factor VIIa/tissue factor, inhibition by AT, interaction profiles with phospholipids, affinities for factor VIIIa within the context of the Xase complex, and thrombin generation profiles were similar for rFIXFc and rFIX. Xase complexes formed with either molecule exhibited similar kinetic profiles for factor Xa generation. In acute efficacy models, mice infused with rFIXFc or rFIX were equally protected from bleeding. However, in prophylactic efficacy models, protection from bleeding was maintained approximately three times longer in rFIXFc-dosed mice than in those given rFIX; this prolonged efficacy correlates with the previously observed half-life extension. We conclude that rFIXFc retains critical FIX procoagulant attributes and that the extension in rFIXFc half-life translates into prolonged efficacy in hemophilia B mice.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Coagulantes/farmacologia , Fator IX/farmacologia , Hemofilia B/tratamento farmacológico , Hemorragia/tratamento farmacológico , Fragmentos Fc das Imunoglobulinas/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Animais , Antitrombina III/farmacologia , Testes de Coagulação Sanguínea , Modelos Animais de Doenças , Ativação Enzimática/fisiologia , Fator IX/genética , Fator VIIa/farmacologia , Fator XIa/farmacologia , Meia-Vida , Fragmentos Fc das Imunoglobulinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/genética , Trombina/biossíntese
8.
J Thromb Haemost ; 13(11): 2041-52, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26340413

RESUMO

BACKGROUND: Conventional coagulation factor assays are associated with certain limitations, as they do not always reflect the clinical heterogeneity of bleeding in hemophilic patients or correctly reflect the individual patient response to treatment with bypassing agents or novel factor concentrates. The thrombin generation assay (TGA) is currently being assessed as a possible method for characterizing bleeding phenotypes in individuals with hemophilia. OBJECTIVES: This study assessed the robustness and sensitivity of the TGA for measuring the activity of recombinant factor VIII (rFVIII), recombinant factor IX (rFIX) and their glycoPEGylated derivatives, N8-GP and N9-GP, in vitro. METHODS: Factor-deficient plasma was spiked with 0.13-130 IU dL(-1) rFVIII or N8-GP (hemophilia A [HA] plasma), or rFIX or N9-GP (hemophilia B [HB] plasma). A calibrated automated thrombogram triggered with tissue factor (TF) or activated FXI (FXIa) was used to measure thrombin generation over time. Endogenous thrombin potential, peak thrombin, velocity index, lag time and time to peak thrombin were analyzed. RESULTS: FXIa-triggered assays were not affected by glycoPEGylation and were sufficiently sensitive to differentiate between spiked samples mimicking severe and moderate HB and HA; TF-triggered assays were not sufficiently sensitive for this distinction in HA. Both FXIa-triggered and TF-triggered assays had an acceptable level of variability (≤ 20%), although TF-triggered assays were associated with greater variability. CONCLUSIONS: FXIa-triggered TGA reactions produced more robust and sensitive results than TF-triggered TGA reactions, and have the potential for use in monitoring patients treated with glycoPEGylated or non-PEGylated coagulation factor concentrates. These promising results merit confirmation with clinical samples to correlate in vitro and in vivo data.


Assuntos
Testes de Coagulação Sanguínea/métodos , Fator IX/análise , Fator VIII/análise , Fator XIa/análise , Hemofilia A/sangue , Polietilenoglicóis/análise , Trombina/biossíntese , Monitoramento de Medicamentos/métodos , Fator VIII/farmacologia , Fator XIa/farmacologia , Humanos , Proteínas Recombinantes/análise , Proteínas Recombinantes/sangue , Proteínas Recombinantes/farmacologia , Sensibilidade e Especificidade , Tromboplastina/farmacologia
9.
Gen Physiol Biophys ; 33(2): 145-55, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24448372

RESUMO

Human plasma contains wide variety of bioactive proteins that have proved essential in therapeutic discovery. However many human plasma proteins remain orphans with unknown biological functions. Evidences suggest that some plasma components target the respiratory system. In the present study we adapted heparin affinity chromatography to fractionate human plasma for functional bioassay. Fractions from pooled human plasma yielded particular plasma fractions with strong cough suppressing effects. Purification yielded a fraction that was finally identified as an activated blood coagulation factor fXIa using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and matrix-assisted laser desorption ionization/time-of-flight mass spectrometry (MALDI/TOF-MS). The fraction almost completely suppressed coughs induced by either chemical or mechanical stimulation applied to larynx or bifurcation of guinea-pig trachea. Cough suppressing effect of the fraction and commercially available fXIa were one million times stronger than codeine and codeine only partially suppressed the mechanically triggered coughing in animal model. Recent reviews highlighted prominent shortcomings of current available antitussives, including narcotic opioids such as codeine and their unpleasant or intolerable side effects. Therefore, safer and more effective cough suppressants would be welcome, and present findings indicate that fXIa in human plasma as a very promising, new therapeutic candidate for effective antitussive action.


Assuntos
Antitussígenos/sangue , Antitussígenos/farmacologia , Tosse/tratamento farmacológico , Animais , Antitussígenos/isolamento & purificação , Antitussígenos/metabolismo , Bioensaio , Análise Química do Sangue , Capsaicina/farmacologia , Cromatografia de Afinidade , Codeína/farmacologia , Tosse/induzido quimicamente , Descoberta de Drogas , Fator XIa/isolamento & purificação , Fator XIa/metabolismo , Fator XIa/farmacologia , Cobaias , Heparina/metabolismo , Masculino , Fenômenos Mecânicos
10.
J Autoimmun ; 39(3): 121-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22704541

RESUMO

Factor XI (FXI), a disulfide-linked covalent homodimer, circulates in plasma, and upon activation initiates the intrinsic/consolidation phase of coagulation. We present evidence that disulfide bonds in FXI are reduced to free thiols by oxidoreductases thioredoxin-1 (TRX-1) and protein disulfide isomerase (PDI). We identified that Cys362-Cys482 and Cys118-Cys147 disulfide bonds are reduced by TRX-1. The activation of TRX-1-treated FXI by thrombin, FXIIa or FXIa was significantly increased compared to non-reduced FXI, indicating that the reduced factor is more efficiently activated than the oxidized protein. Using a novel ELISA system, we compared the amount of reduced FXI in antiphospholipid syndrome (APS) thrombosis patients with levels in healthy controls, and found that APS patients have higher levels of reduced FXI. This may have implication for understanding the contribution of FXI to APS thrombosis, and the predisposition to thrombosis in patients with elevated plasma levels of reduced FXI.


Assuntos
Síndrome Antifosfolipídica/sangue , Fator XI/agonistas , Isomerases de Dissulfetos de Proteínas/sangue , Tiorredoxinas/sangue , Trombose/sangue , Adulto , Idoso , Síndrome Antifosfolipídica/complicações , Síndrome Antifosfolipídica/enzimologia , Coagulação Sanguínea , Estudos de Casos e Controles , Cisteína/metabolismo , Dissulfetos/química , Ensaio de Imunoadsorção Enzimática , Fator XI/química , Fator XI/metabolismo , Fator XIIa/metabolismo , Fator XIIa/farmacologia , Fator XIa/metabolismo , Fator XIa/farmacologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Oxirredução , Tiorredoxinas/metabolismo , Tiorredoxinas/farmacologia , Trombina/metabolismo , Trombina/farmacologia , Trombose/complicações , Trombose/enzimologia
11.
J Leukoc Biol ; 90(5): 923-7, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21807745

RESUMO

PMN leukocytes are the most abundant leukocytes in the circulation and play an important role in host defense. PMN leukocyte recruitment and inflammatory responses at sites of infection are critical components in innate immunity. Although inflammation and coagulation are known to have bidirectional relationships, little is known about the interaction between PMN leukocytes and coagulation factors. Coagulation FXI participates in the intrinsic coagulation pathway upon its activation, contributing to hemostasis and thrombosis. We have shown previously that FXI-deficient mice have an increased survival and less leukocyte accumulation into the peritoneum in severe polymicrobial peritonitis. This result suggests a role for FXI in leukocyte trafficking and/or function. In this study, we characterized the functional consequences of FXIa binding to PMN leukocytes. FXIa reduced PMN leukocyte chemotaxis triggered by the chemokine, IL-8, or the bacterial-derived peptide, fMLP, perhaps as a result of the loss of directed migration. In summary, our data suggest that FXIa modulates the inflammatory response of PMN leukocytes by altering migration. These studies highlight the interplay between inflammation and coagulation and suggest that FXIa may play a role in innate immunity.


Assuntos
Quimiotaxia de Leucócito/fisiologia , Fator XIa/metabolismo , Neutrófilos/fisiologia , Coagulação Sanguínea , Movimento Celular , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Fator XIa/farmacologia , Humanos , Inflamação/metabolismo , Interleucina-8/farmacologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/efeitos dos fármacos , Peritonite/metabolismo , Ligação Proteica
12.
Blood ; 101(12): 4783-8, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12586617

RESUMO

Factor XI deficiency, an injury-related bleeding disorder, is rare worldwide but common in Jews in whom 2 mutations, Glu117Stop (type II) and Phe283Leu (type III), prevail. Mean factor XI activities in homozygotes for Glu117Stop and for Phe283Leu are 1 and 10 U/dL, respectively. Inhibitors to factor XI in patients with severe factor XI deficiency have been reported in a small number of instances. This study was undertaken to determine the prevalence of acquired inhibitors against factor XI in patients with severe factor XI deficiency, discern whether these inhibitors are related to specific mutations, and characterize their activity. Clinical information was obtained from unrelated patients with severe factor XI deficiency, and blood was analyzed for factor XI activity, inhibitor to factor XI, and causative mutations. Immunoglobulin G purified from patients with an inhibitory activity was tested for binding to factor XI, effects on activation of factor XI by factor XIIa and thrombin, and activation of factor IX by exogenous factor XIa. Of 118 Israeli patients, 7 had an inhibitor; all belonged to a subgroup of 21 homozygotes for Glu117Stop who had a history of plasma replacement therapy. Three additional patients with inhibitors from the United Kingdom and the United States also had this genotype and were exposed to plasma. The inhibitors affected factor XI activation by thrombin or factor XIIa, and activation of factor IX by factor XIa. The results imply that patients with a very low factor XI level are susceptible to development of an inhibitor following plasma replacement.


Assuntos
Deficiência do Fator XI/epidemiologia , Deficiência do Fator XI/etiologia , Idoso , Autoanticorpos/sangue , Fator IX/metabolismo , Fator VIIa/farmacologia , Fator XI/imunologia , Fator XI/metabolismo , Deficiência do Fator XI/genética , Deficiência do Fator XI/imunologia , Fator XIIa/farmacologia , Fator XIa/farmacologia , Feminino , Genótipo , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Imunoglobulina G/sangue , Israel , Judeus , Masculino , Pessoa de Meia-Idade , Mutação , Tempo de Tromboplastina Parcial , Plasma , Proteínas Recombinantes/farmacologia , Trombina/metabolismo , Trombina/farmacologia , Reino Unido , Estados Unidos
13.
J Biol Chem ; 275(27): 20514-9, 2000 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-10781579

RESUMO

To study the pathways for initiation of intrinsic blood coagulation, activated human platelets were compared with dextran sulfate as surfaces for factor XI activation by factor XIIa, factor XIa, or thrombin. Activated gel-filtered platelets promoted the activation of factor XI (60 nm) by thrombin (0.02-10 nm, EC(50) approximately 100 pm, threshold concentration approximately 10 pm) at initial rates 2- to 3-fold greater than those obtained with dextran sulfate in the presence of either high molecular weight kininogen (45 nm) and ZnCl(2) (25 micrometer) or prothrombin (1.2 micrometer) and CaCl(2) (2 mm). The maximum rates of factor XI activation achieved in the presence of activated gel-filtered platelets were 30 nm.min(-1) with thrombin, 6 nm.min(-1) with factor XIIa and 2 nm.min(-1) with factor XIa. Values of turnover number calculated at various enzyme concentrations (0.05-1 nm) were 24-167 (mean = 86) min(-1) for thrombin, 4.6-50 (mean = 21) min(-1) for factor XIIa, and 1.3-14 (mean = 8) min(-1) for factor XIa. A physiological concentration of fibrinogen (9.0 micrometer) inhibited factor XI activation by thrombin (but not by factor XIIa) in the presence of dextran sulfate but not in the presence of gel-filtered platelets. Compared with factors XIIa and XIa, thrombin is the preferred factor XI activator, and activated platelets are a relevant physiological surface for thrombin-mediated initiation of intrinsic coagulation in vivo.


Assuntos
Plaquetas/metabolismo , Fator XIIa/farmacologia , Fator XI/metabolismo , Fator XIa/farmacologia , Ativação Plaquetária , Trombina/farmacologia , Plaquetas/enzimologia , Sulfato de Dextrana/farmacologia , Fibrinogênio/farmacologia , Humanos , Propriedades de Superfície
14.
J Biol Chem ; 272(37): 23418-26, 1997 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-9287357

RESUMO

We previously identified a high affinity Ca2+ binding site in the protease domain of factor IXa involving Glu235 (Glu70 in chymotrypsinogen numbering; hereafter, the numbers in brackets refer to the chymotrypsin equivalents) and Glu245[80] as putative ligands. To delineate the function of this Ca2+ binding site, we expressed IXwild type (IXWT), IXE235K, and IXE245V in 293 kidney cells and compared their properties with those of factor IX isolated from normal plasma (IXNP); each protein had the same Mr and gamma-carboxyglutamic acid content. Activation of each factor IX protein by factor VIIa.Ca2+.tissue factor was normal as analyzed by sodium dodecyl sulfate-gel electrophoresis. The coagulant activity of IXaWT was approximately 93%, of IXaE235K was approximately 27%, and of IXaE245V was approximately 4% compared with that of IXaNP. In contrast, activation by factor XIa.Ca2+ led to proteolysis at Arg318-Ser319[150-151] in the protease domain autolysis loop of IXaE245V with a concomitant loss of coagulant activity; this proteolysis was moderate in IXaE235K and minimal in IXaWT or IXaNP. Interaction of each activated mutant with an active site probe, p-aminobenzamidine, was also examined; the Kd of interaction in the absence and presence (in parentheses) of Ca2+ was: IXaNP or IXaWT 230 microM (78 microM), IXaE235K 150 microM (145 microM), IXaE245V 225 microM (240 microM), and autolysis loop cleaved IXaE245V 330 microM (350 microM). Next, we evaluated the apparent Kd (Kd,app) of interaction of each activated mutant with factor VIIIa. We first investigated the EC50 of interaction of IXaNP as well as of IXaWT with factor VIIIa in the presence and absence of phospholipid (PL) and varying concentrations of factor X. At each factor X concentration and constant factor VIIIa, EC50 was the free IXaNP or IXaWT concentration that yielded a half-maximal rate of factor Xa generation. EC50 values for IXaNP and IXaWT were similar and are as follows: PL-minus/X-minus (extrapolated), 2.8 microM; PL-minus/X-saturating, 0.25 microM; PLplus/X-minus, 1.6 nM; and PL-plus/X-saturating, 0.09 nM. Further, Kd,app of binding of active site-blocked factor IXa to factor VIIIa was calculated from its ability to inhibit IXaWT in the Tenase assay. Kd,app values in the absence and presence (in parentheses) of PL were: IXaNP or IXaWT, 0. 19 microM (0.07 nM); IXaE235K, 0.68 microM (0.26 nM); IXaE245V, 2.5 microM (1.35 nM); and autolysis loop-cleaved IXaE245V, 15.6 microM (14.3 nM). We conclude that (a) PL increases the apparent affinity of factor IXa for factor VIIIa approximately 2,000-fold, and the substrate, factor X, increases this affinity approximately 10-15-fold; (b) the protease domain Ca2+ binding site increases this affinity approximately 15-fold, and lysine at position 235 only partly substitutes for Ca2+; (c) Ca2+ binding to the protease domain increases the S1 reactivity approximately 3-fold and prevents proteolysis in the autolysis loop; and (d) proteolysis in the autolysis loop leads to a loss of catalytic efficiency with retention of S1 binding site and a further approximately 8-fold reduction in affinity of factor IXa for factor VIIIa.


Assuntos
Fator IXa/metabolismo , Fator VIIIa/metabolismo , Ácido 1-Carboxiglutâmico/análise , Benzamidinas/metabolismo , Sítios de Ligação , Ligação Competitiva , Cálcio/metabolismo , Ativação Enzimática , Fator IXa/genética , Fator VIIa/farmacologia , Fator X/farmacologia , Fator XIa/farmacologia , Modelos Químicos , Modelos Moleculares , Mutação , Fragmentos de Peptídeos/farmacologia , Fosfolipídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Análise de Sequência
15.
J Clin Invest ; 100(4): 886-92, 1997 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-9259588

RESUMO

Using the techniques of molecular biology, we made a chimeric Factor IX by replacing the first epidermal growth factor-like domain with that of Factor VII. The resulting recombinant chimeric molecule, Factor IXVIIEGF1, had at least a twofold increase in functional activity in the one-stage clotting assay when compared to recombinant wild-type Factor IX. The increased activity was not due to contamination with activated Factor IX, nor was it due to an increased rate of activation by Factor VIIa-tissue factor or by Factor XIa. Rather, the increased activity was due to a higher affinity of Factor IXVIIEGF1 for Factor VIIIa with a Kd for Factor VIIIa about one order of magnitude lower than that of recombinant wild-type Factor IXa. In addition, results from animal studies show that this chimeric Factor IX, when infused into a dog with hemophilia B, exhibits a greater than threefold increase in clotting activity, and has a biological half-life equivalent to recombinant wild-type Factor IX.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Fator IX/metabolismo , Hemofilia B/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , DNA Recombinante/metabolismo , Compostos de Dansil/farmacologia , Cães , Fator de Crescimento Epidérmico/genética , Fator IX/genética , Fator IX/imunologia , Fator IX/isolamento & purificação , Fator IXa/metabolismo , Fator VIIIa/metabolismo , Fator VIIa/farmacologia , Fator XIa/farmacologia , Fator Xa/metabolismo , Inibidores do Fator Xa , Humanos , Tempo de Tromboplastina Parcial , Proteínas Recombinantes de Fusão/isolamento & purificação
16.
Thromb Haemost ; 75(3): 445-9, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8701405

RESUMO

Coagulation factor XI is a glycoprotein of the contact factor system. Its deficiency is associated with a highly variable bleeding tendency, thus a role in relation to hemostasis appears to exist. However, the importance of factor XI for stimulating intrinsic coagulation in vivo has not yet been determined. To study the procoagulant effects of human factor XIa in vivo, we infused the purified enzyme into normal chimpanzees (100 micrograms) in the absence or presence of the thrombin inhibitor rec-hirudin (1.0 mg/kg loading dose plus 0.3 mg/kg body wt continuous infusion). Factor XIa elicited an immediate activation of factors IX, X, and prothrombin, as measured by their respective activation fragments. However, whereas the activation of factors IX and X was immediate and shortlasting, (peak increments of 6- and 1.4-fold of baseline at 5 minutes after injection), the conversion of prothrombin gradually increased, reaching a summit of 6-fold baseline values after 60 min, and remaining elevated during the course of the experiments. Thrombin-antithrombin complexes also remained elevated during the study period. In the presence of hirudin, the initial activation of factors IX, X, and prothrombin was unchanged, however the further increment in prothrombin fragment F1 + 2 was markedly inhibited. These results demonstrate that factor XIa is a potential agonist of the intrinsic cascade in vivo, which activity is enhanced in the presence of thrombin.


Assuntos
Fatores de Coagulação Sanguínea/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Fator XIa/farmacologia , Análise de Variância , Animais , Biomarcadores/sangue , Humanos , Técnicas In Vitro , Pan troglodytes
17.
Biochemistry ; 33(47): 14136-43, 1994 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-7947825

RESUMO

The gamma-carboxylase recognition site in the propeptide of profactor IX signals the gamma-carboxylation of specific glutamic acid residues in the adjacent Gla domain during factor IX biosynthesis. To study posttranslational processing of the vitamin K-dependent blood coagulation factors and the properties of processing intermediates, we have isolated an incompletely processed factor IX species, profactor IX, from the medium of heterologous mammalian cells expressing the human factor IX cDNA. Profactor IX was purified by sequential immunoaffinity chromatography using antibodies specific for the propeptide and antibodies specific for the well-carboxylated factor IX species. This purified profactor IX preparation was fully gamma-carboxylated and contained the N-terminal propeptide, but it exhibited no factor IX procoagulant activity. Profactor IX was not cleaved following incubation with factor XIa. In contrast to mature factor IX, profactor IX did not demonstrate Ca(II)-dependent binding to acidic phospholipid vesicles, nor can the membrane binding surface be expressed, as detected by antibodies specific for this epitope. The propeptide of profactor IX can be removed in vitro by a specific endopeptidase, furin/PACE, yielding factor IX, which can be converted to fully active factor IXa by factor XIa and which binds normally to acidic phospholipid vesicles. These results indicate that fully gamma-carboxylated profactor IX is biologically inactive due to the presence of the propeptide.


Assuntos
Membrana Celular/metabolismo , Fator IX/farmacologia , Fator XIa/farmacologia , Precursores de Proteínas/farmacologia , Animais , Sítios de Ligação , Western Blotting , Células CHO , Cálcio/farmacologia , Carboxiliases/metabolismo , Cricetinae , Ácido Edético/farmacologia , Fator IX/isolamento & purificação , Fator IX/metabolismo , Humanos , Lipossomos/metabolismo , Fosfolipídeos/metabolismo , Conformação Proteica , Precursores de Proteínas/isolamento & purificação , Precursores de Proteínas/metabolismo , Proteínas Recombinantes/isolamento & purificação
18.
Blood ; 84(6): 1866-73, 1994 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-7915915

RESUMO

Factor IXTaipei9 is a factor IX variant from a hemophilia B patient with reduced levels of circulating protein molecules (cross-reacting material reduced, CRM). This variant contained a glycine (Gly) to glutamic acid (Glu) substitution at the 207th codon of mature factor IX. The functional consequences of the Gly-->Glu mutation in factor IXTaipei9 (IXG207E) were characterized in this study. Plasma-derived IXG207E exhibited a mobility similar to that of normal factor IX on sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Its specific activity was estimated to be 3.5% that of the purified normal factor IX in a one-stage partial thromboplastin time assay (aPTT). Cleavage of factor IXG207E by factor XIa or factor VIIa-tissue factor complex appeared to be normal. When the calcium-dependent conformational change was examined by monitoring quenching of intrinsic fluorescence, both normal factor IX and IXG207E exhibited equivalent intrinsic fluorescence quenching. Activated factor IXG207E (IXaG207E) also binds antithrombin III equally as well as normal factor IXa. However, aberrant binding of the active site probe p-aminobenzamidine was observed for factor XIa-activated factor IXG207E, indicating that the active site pocket of the heavy chain of factor IXaG207E was abnormal. Moreover, the rate of activation of factor X by factor IXaG207E, as measured in a purified system using chromogenic substrates, was estimated to be 1/40 of that of normal factor IXa. A computer-modeled heavy-chain structure of factor IXa predicts a hydrophobic environment surrounding Gly-207 and this Gly forms a hydrogen bound to the active site serine-365. The molecular mechanism of the Gly-->Glu mutation in factor IXTaipei9 might result in the alteration of the microenvironment of the active site pocket which renders the active site serine-365 inaccessible to its substrate.


Assuntos
Fator IX/genética , Glutamatos/genética , Glicina/genética , Hemofilia B/genética , Mutação , Benzamidinas/metabolismo , Sítios de Ligação , Cálcio/farmacologia , Códon , Simulação por Computador , Eletroforese em Gel de Poliacrilamida , Fator IX/química , Fator IX/metabolismo , Fator VIIa/metabolismo , Fator X/metabolismo , Fator XIa/farmacologia , Ácido Glutâmico , Humanos , Masculino , Modelos Moleculares , Tempo de Tromboplastina Parcial , Taiwan
19.
Blood ; 76(3): 549-54, 1990 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-2165828

RESUMO

Generation and inhibition of activated factor IXa was studied in factor XIa-activated plasma containing 4 mmol/L free calcium ions and 20 mumol/L phospholipid (25 mol% phosphatidylserine/75 mol% phosphatidylcholine). Interference of other (activated) clotting factors with the factor IXa activity measurements could be avoided by using a highly specific and sensitive bioassay. Factor IXa generation curves were analyzed according to a model that assumed Michaelis-Menten kinetics of factor XIa-catalyzed factor IXa formation and pseudo first order kinetics of inhibition of factor XIa and factor IXa. In the absence of heparin, factor IXa activity in plasma reached final levels that were found to increase with increasing amounts of factor XIa used to activate the plasma. When the model was fitted to this set of factor IXa generation curves, the analysis yielded a rate constant of inhibition of factor XIa of 0.7 +/- 0.1 min-1 and a kcat/Km ratio of 0.29 +/- 0.01 (nmol/L)-1 min-1. No neutralization of factor IXa activity was observed (the estimated rate constant of inhibition of factor IXa was 0). Thus, in the absence of heparin, the final level of factor IXa in plasma is only dependent on the initial factor XIa concentration. While neutralization of in situ generated factor IXa in normal plasma was negligible, unfractionated heparin dramatically enhanced the rate of inactivation of factor IXa (apparent second order rate constant of inhibition of 5.2 min-1/per microgram heparin/mL). The synthetic pentasaccharide heparin, the smallest heparin chain capable of binding antithrombin III, stimulated the inhibition of in situ generated factor IXa, but sevenfold less than unfractionated heparin (k = 0.76 min-1 per microgram pentasaccharide/mL). We found that free calcium ions were absolutely required to observe an unfractionated heparin and pentasaccharide-stimulated neutralization of factor IXa activity. Factor XIa inhibition (psuedo first order rate constant of 0.7 min-1) was not affected by unfractionated heparin or pentasaccharide in the range of heparin concentrations studied.


Assuntos
Fator IXa/metabolismo , Heparina/farmacologia , Análise por Ativação , Antitrombina III/metabolismo , Bioensaio , Fator XIa/metabolismo , Fator XIa/farmacologia , Heparina/análise , Heparina/metabolismo , Heparina de Baixo Peso Molecular/farmacologia , Humanos , Peso Molecular
20.
J Biol Chem ; 265(1): 144-50, 1990 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-2294100

RESUMO

Human blood clotting factor IX, and two chimeric molecules of factor IX, in which the first epidermal growth factor-like domain or both epidermal growth factor-like domains have been replaced by that of human factor X, have been expressed in mouse C127 cells. The recombinants have been purified using a metal ion-dependent monoclonal antibody specific for residues 1-42 of human factor IX. All recombinant molecules are activated normally by human factor XIa in the presence of calcium ion. Activation of the factor IX recombinants by factor VIIa-tissue factor appears to be normal for the epidermal growth factor-1 exchange but considerably reduced for the construction containing both epidermal growth factor-like domains of factor X. The analysis of gamma-carboxyglutamic acid residues reveals that all of the purified recombinants are almost fully carboxylated. The extent of aspartic acid hydroxylation at residue 64 is 60% for all recombinants. The chimeric molecule with both epidermal growth factor-like domains from factor X has about 4% normal activity in the activated partial thromboplastin time assay. In contrast, the construct containing the first epidermal growth factor-like domain of factor X shows essentially normal clotting activity. Thus, it is unlikely that this domain is involved in a unique interaction with factor VIII.


Assuntos
Fator de Crescimento Epidérmico/genética , Fator IX/genética , Fator X/genética , Expressão Gênica , Proteínas Recombinantes/genética , Ácido 1-Carboxiglutâmico/análise , Sequência de Aminoácidos , Animais , Ácido Aspártico/metabolismo , Sequência de Bases , Coagulação Sanguínea , Cálcio/farmacologia , Linhagem Celular , Fator IX/fisiologia , Fator VIIa/farmacologia , Fator X/fisiologia , Fator XIa/farmacologia , Humanos , Hidroxilação , Camundongos , Dados de Sequência Molecular , Proteínas Recombinantes/análise , Proteínas Recombinantes/fisiologia , Tromboplastina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...