Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 470
Filtrar
1.
Int J Mol Sci ; 24(5)2023 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-36902328

RESUMEN

Direct FXa inhibitors are an important class of bioactive molecules (rivaroxaban, apixaban, edoxaban, and betrixaban) applied for thromboprophylaxis in diverse cardiovascular pathologies. The interaction of active compounds with human serum albumin (HSA), the most abundant protein in blood plasma, is a key research area and provides crucial information about drugs' pharmacokinetics and pharmacodynamic properties. This research focuses on the study of the interactions between HSA and four commercially available direct oral FXa inhibitors, applying methodologies including steady-state and time-resolved fluorescence, isothermal titration calorimetry (ITC), and molecular dynamics. The HSA complexation of FXa inhibitors was found to occur via static quenching, and the complex formation in the ground states affects the fluorescence of HSA, with a moderate binding constant of 104 M-1. However, the ITC studies reported significantly different binding constants (103 M-1) compared with the results obtained through spectrophotometric methods. The suspected binding mode is supported by molecular dynamics simulations, where the predominant interactions were hydrogen bonds and hydrophobic interactions (mainly π-π stacking interactions between the phenyl ring of FXa inhibitors and the indole moiety of Trp214). Finally, the possible implications of the obtained results regarding pathologies such as hypoalbuminemia are briefly discussed.


Asunto(s)
Factor X , Albúmina Sérica Humana , Tromboembolia Venosa , Humanos , Anticoagulantes , Sitios de Unión , Calorimetría/métodos , Simulación del Acoplamiento Molecular , Unión Proteica , Albúmina Sérica Humana/química , Espectrometría de Fluorescencia , Termodinámica , Factor X/antagonistas & inhibidores
2.
Blood ; 138(14): 1258-1268, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34077951

RESUMEN

Hemophilia A is a bleeding disorder resulting from deficient factor VIII (FVIII), which normally functions as a cofactor to activated factor IX (FIXa) that facilitates activation of factor X (FX). To mimic this property in a bispecific antibody format, a screening was conducted to identify functional pairs of anti-FIXa and anti-FX antibodies, followed by optimization of functional and biophysical properties. The resulting bispecific antibody (Mim8) assembled efficiently with FIXa and FX on membranes, and supported activation with an apparent equilibrium dissociation constant of 16 nM. Binding affinity with FIXa and FX in solution was much lower, with equilibrium dissociation constant values for FIXa and FX of 2.3 and 1.5 µM, respectively. In addition, the activity of Mim8 was dependent on stimulatory activity contributed by the anti-FIXa arm, which enhanced the proteolytic activity of FIXa by 4 orders of magnitude. In hemophilia A plasma and whole blood, Mim8 normalized thrombin generation and clot formation, with potencies 13 and 18 times higher than a sequence-identical analogue of emicizumab. A similar potency difference was observed in a tail vein transection model in hemophilia A mice, whereas reduction of bleeding in a severe tail-clip model was observed only for Mim8. Furthermore, the pharmacokinetic parameters of Mim8 were investigated and a half-life of 14 days shown in cynomolgus monkeys. In conclusion, Mim8 is an activated FVIII mimetic with a potent and efficacious hemostatic effect based on preclinical data.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Hemofilia A/tratamiento farmacológico , Hemorragia/tratamiento farmacológico , Animales , Factor IXa/antagonistas & inhibidores , Factor VIIIa/uso terapéutico , Factor X/antagonistas & inhibidores , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL
3.
Thromb Haemost ; 120(11): 1512-1523, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32820486

RESUMEN

The venom of the Australian snake Pseudonaja textilis comprises powerful prothrombin activators consisting of factor X (v-ptFX)- and factor V-like proteins. While all vertebrate liver-expressed factor X (FX) homologs, including that of P. textilis, comprise an activation peptide of approximately 45 to 65 residues, the activation peptide of v-ptFX is significantly shortened to 27 residues. In this study, we demonstrate that exchanging the human FX activation peptide for the snake venom ortholog impedes proteolytic cleavage by the intrinsic factor VIIIa-factor IXa tenase complex. Furthermore, our findings indicate that the human FX activation peptide comprises an essential binding site for the intrinsic tenase complex. Conversely, incorporation of FX into the extrinsic tissue factor-factor VIIa tenase complex is completely dependent on exosite-mediated interactions. Remarkably, the shortened activation peptide allows for factor V-dependent prothrombin conversion while in the zymogen state. This indicates that the active site of FX molecules comprising the v-ptFX activation peptide partially matures upon assembly into a premature prothrombinase complex. Taken together, the shortened activation peptide is one of the remarkable characteristics of v-ptFX that has been modified from its original form, thereby transforming FX into a powerful procoagulant protein. Moreover, these results shed new light on the structural requirements for serine protease activation and indicate that catalytic activity can be obtained without formation of the characteristic Ile16-Asp194 salt bridge via modification of the activation peptide.


Asunto(s)
Venenos Elapídicos/metabolismo , Elapidae/metabolismo , Factor X/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Sitios de Unión , Unión Competitiva , Catálisis , Dominio Catalítico , Cisteína Endopeptidasas , Venenos Elapídicos/genética , Activación Enzimática , Evolución Molecular , Factor VIIIa/metabolismo , Factor VIIa/metabolismo , Factor X/antagonistas & inhibidores , Factor X/genética , Humanos , Complejos Multiproteicos , Fragmentos de Péptidos/farmacología , Pirazoles/farmacología , Piridonas/farmacología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Tromboplastina/metabolismo
4.
Hematology Am Soc Hematol Educ Program ; 2019(1): 194-197, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31808890

RESUMEN

Direct oral anticoagulants (DOACs) are increasingly used in the treatment and prophylaxis of thromboembolism because of several advantages over vitamin K antagonists, including no need for laboratory monitoring. However, it has become increasingly important in certain clinical scenarios to know either actual DOAC concentration (quantitative) or presence of DOAC (qualitative). These clinical conditions include patients presenting with major bleeding or requiring urgent surgery who may need a reversal or hemostatic agent, extremes of body weight, failed therapy, etc. Prothrombin time and activated partial thromboplastin time are variably affected by factor Xa inhibitors (FXaIs) and direct thrombin inhibitor (DTI), respectively, depending on reagents' sensitivity, and hence, they cannot be relied on confidently. Thrombin time is highly sensitive to very low amounts of DTI; thus, normal value rules out a clinically significant amount. Liquid chromatography mass spectrometry accurately measures DOAC levels but is clinically impractical. Dilute thrombin time and ecarin-based assays using appropriate calibrators/controls provide an accurate DTI level. Anti-Xa assay using corresponding FXaI calibrators/controls provides accurate drug levels. However, these assays are not readily available in the United States compared with some other parts of the world. Heparin assays using anti-Xa activity often have a linear relationship with calibrated FXaI assays, especially at the lower end of on-therapy levels, and they may provide rapid assessment of drug activity for clinical decision making. Currently, there is very limited knowledge of DOAC effect on viscoelastic measurements. Although there is uniformity in expression of DOAC concentrations in nanograms per milliliter, a universal FXaI DOAC assay is urgently needed.


Asunto(s)
Anticoagulantes/administración & dosificación , Anticoagulantes/uso terapéutico , Técnicas de Laboratorio Clínico , Monitoreo de Drogas , Administración Oral , Anciano de 80 o más Años , Factor X/antagonistas & inhibidores , Factor X/metabolismo , Femenino , Humanos
6.
Hosp Pract (1995) ; 47(3): 113-122, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31317796

RESUMEN

Direct oral anticoagulants (DOACs) include dabigatran etexilate, a direct thrombin inhibitor, and specific inhibitors of activated coagulation factor X (FXa; e.g. apixaban, betrixaban, edoxaban, rivaroxaban). DOACs are associated with lower rates of major and fatal bleeding events compared with warfarin. Clinicians may need to achieve rapid reversal of anticoagulation effects of the DOACs in an emergency setting. Idarucizumab and andexanet alfa, which reverse the anticoagulant effects of dabigatran and FXa inhibitors, respectively, are DOAC reversal agents available in the US. Other reversal agents (e.g. ciraparantag for heparins, DOACs) are in development. Alternative nonspecific agents (e.g. fresh frozen plasma, prothrombin complex concentrate) are available. Nonspecific prohemostatic agents can counteract the anticoagulant action of DOACs in emergency situations, when specific reversal agents are unavailable. However, specific reversal agents are efficacious and safe and should be preferred when available. In this review, we discuss practical issues in the initiation of DOAC therapy, situations where reversal may be needed, coagulation assays, reversal agents, and post-reversal complications in the context of published evidence and guidelines.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticoagulantes/administración & dosificación , Antitrombinas/uso terapéutico , Dabigatrán/antagonistas & inhibidores , Factor X/antagonistas & inhibidores , Factor Xa/farmacología , Pacientes Internos , Proteínas Recombinantes/farmacología , Administración Oral , Coagulación Sanguínea/efectos de los fármacos , Servicios Médicos de Urgencia , Factor Xa/agonistas , Inhibidores del Factor Xa/uso terapéutico , Hemorragia/tratamiento farmacológico , Humanos , Cuerpo Médico de Hospitales
7.
Chem Biol Drug Des ; 94(3): 1664-1671, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31108011

RESUMEN

Rivaroxaban (RIV) is a direct oral anticoagulant (DOAC) targeting activated coagulation factor X (FXa). An earlier study reported the F174A mutant of FXa resistant to a RIV-like inhibitor, Apixaban. In current study, the detailed molecular mechanism of the resistance has been explored by molecular dynamics simulations on the impaired interactions between RIV and FXa in the damaged S4 pocket of F174A mutant. Besides, an unexpected relative stable binding mode of S1'S1 was revealed, which required dynamic motions of Gln192 and Gln61 to allow the morpholinone moiety of RIV to shift into the S1' pocket and form strong interactions. These dynamic motions of RIV and critical residues might be important in drug design for direct inhibitors of coagulation factors.


Asunto(s)
Anticoagulantes/química , Factor X/antagonistas & inhibidores , Inhibidores del Factor Xa/química , Simulación de Dinámica Molecular , Proteínas Mutantes/antagonistas & inhibidores , Pirazoles/química , Piridonas/química , Rivaroxabán/química , Secuencia de Aminoácidos , Sitios de Unión , Diseño de Fármacos , Factor X/genética , Humanos , Proteínas Mutantes/genética , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
9.
J Thromb Haemost ; 17(1): 126-137, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30444568

RESUMEN

Essentials Emicizumab mimics factor (F)VIIIa cofactor function, augments the intrinsic tenase activity. We assessed the emicizumab-driven hemostatic function in FXI-deficient plasmas. Emicizumab improved the coagulation potentials in severe FXI-deficient plasma. Emicizumab may provide a possibility for clinical application in patients with FXI deficiency. SUMMARY: Background Patients with factor (F)XI deficiency commonly present with markedly prolonged activated partial thromboplastin times (APTT), although bleeding phenotypes are heterogeneous. Emicizumab, a bispecific monoclonal antibody to FIX/FIXa and FX/FXa, mimics FVIIIa cofactor function on phospholipid (PL) surfaces. Antibody reactions were designed, therefore, to augment mechanisms during the propagation phase of blood coagulation. Aim To assess emicizumab-driven hemostatic function in FXI-deficient plasmas. Methods and Results Standard ellagic acid (Elg)/PL-based APTTs of different FXI-deficient plasmas (n = 13; FXI activity, < 1 IU dl-1 ) were markedly shortened dose dependently by the presence of emicizumab. To further analyze the effects of emicizumab, clot waveform analysis (CWA) in FXI-deficient plasmas with emicizumab, triggered by tissue factor (TF)/Elg demonstrated improvements in both clot times, reflecting the initiation phase, and coagulation velocity, which represents the propagation phase. Emicizumab also enhanced the TF/Elg-triggered thrombin generation in FXI-deficient plasmas dose-dependently although the degree of enhancement varied in individual cases. Thrombin generation with either FVII-deficient plasma or FIX-deficient plasma treated with anti-FXI antibody showed little or no increase by the co-presence of emicizumab, suggesting that the accelerated thrombin generation in FXI-deficient plasmas by emicizumab should depend on the FIXa-involved coagulation propagation initially triggered by FVIIa/TF. The ex vivo addition of emicizumab to whole blood from three patients with severe FXI deficiency demonstrated modest, dose-dependent improvements in Ca2+ -triggered thromboelastograms (NATEM mode). Conclusion Emicizumab appeared to improve coagulation function in severe FXI-deficient plasma, and might provide possibilities for clinical application in patients with FXI deficiency.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Coagulación Sanguínea/efectos de los fármacos , Coagulantes/farmacología , Factor IX/antagonistas & inhibidores , Factor X/antagonistas & inhibidores , Inhibidores del Factor Xa/farmacología , Hemofilia B/tratamiento farmacológico , Estudios de Casos y Controles , Factor IX/metabolismo , Factor X/metabolismo , Factor XIa/antagonistas & inhibidores , Factor XIa/metabolismo , Factor Xa/metabolismo , Hemofilia B/sangre , Humanos , Tiempo de Tromboplastina Parcial , Índice de Severidad de la Enfermedad , Tromboelastografía , Trombina/metabolismo
10.
Structure ; 26(9): 1178-1186.e3, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30017565

RESUMEN

Despite being initially identified in the blood filtrate, LEKTI is a 15-domain Kazal-type inhibitor mostly known in the regulation of skin desquamation. In the current study, screening of serine proteases in blood coagulation cascade showed that LEKTI domain 4 has inhibitory activity toward only FXIa, whereas LEKTI domain 6 inhibits both FXIa and FXaB (bovine FXa). Nuclear magnetic resonance structural and dynamic experiments plus molecular dynamics simulation revealed that LEKTI domain 4 has enhanced backbone flexibility at the reactive-site loop. A model of the LEKTI-protease complex revealed that FXaB has a narrower S4 pocket compared with FXIa and hence prefers only small side-chain residues at the P4 position, such as Ala in LEKTI domain 6. Mutational studies combined with a molecular complex model suggest that both a more flexible reactive-site loop and a bulky residue at the P4 position make LEKTI domain 4 a weaker but highly selective inhibitor of FXIa.


Asunto(s)
Factor XI/antagonistas & inhibidores , Factor X/antagonistas & inhibidores , Inhibidor de Serinpeptidasas Tipo Kazal-5/química , Inhibidor de Serinpeptidasas Tipo Kazal-5/metabolismo , Animales , Sitios de Unión , Coagulación Sanguínea , Bovinos , Factor X/química , Factor XI/química , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Inhibidor de Serinpeptidasas Tipo Kazal-5/genética , Especificidad por Sustrato
11.
J Thromb Haemost ; 16(6): 1078-1088, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29645406

RESUMEN

Essentials The activated partial prothrombin time (aPTT) cannot predict the activity of emicizumab (Emi). Adjusted clot waveform analyses using a prothrombin time (PT)/aPTT initiator were developed. Activity of Emi in the co-presence of factor VIII or bypassing agents was quantified. This assay is useful for assessing coagulation potential in Emi-treated hemophilia A. SUMMARY: Background Emicizumab is an anti-activated factor IX/FX bispecific antibody that mimics activated FVIII cofactor function. Emicizumab does not require activation by thrombin, and its effect on shortening the activated partial thromboplastin time (APTT) is much greater than that of FVIII. Therefore, the APTT has limited utility in hemophilia A (HA) patients treated with emicizumab. Aim To evaluate the global coagulation potential of emicizumab. Methods Clot waveform analysis (CWA) with prothrombin time (PT)/APTT mixed reagents was used to define hemostatic monitoring protocols in HA patients. A modified parameter, adjusted-|min1| (Ad|min1|), was developed. Maximum and minimum percentage transmittance were defined as 100% and 0% in the precoagulation and postcoagulation phases, respectively. Ad|min1| was calculated as an index of the maximum velocity of the coagulation process. Results Ad|min1| obtained with mixed-trigger reagent (PT/APTT/buffer, 1 : 15 : 135) in the presence of emicizumab optimally corresponded to the conversion rate estimated in animals; 0.2-0.4 IU dL-1 equivalent FVIII per 1 µg mL-1 emicizumab). Ex vivo addition of emicizumab to HA plasma with or without inhibitors resulted in concentration-dependent increases in Ad|min1|, with some individual variations. The addition of various concentrations of FVIII to HA plasma mixed with emicizumab resulted in dose-dependent increases in Ad|min1|. Similarly, mixtures of activated prothrombin complex concentrate and emicizumab added to HA plasma resulted in dose-dependent increases in Ad|min1|. In contrast, enhanced coagulation potential appeared to be better defined by the clot time than by Ad|min1| in experiments using recombinant activated FVII. Conclusion The PT/APTT reagent-triggered adjusted CWA could provide a useful means of assessing global coagulation potential in emicizumab-treated HA patients, with enhanced activity neither masking nor being masked by FVIII or bypassing agents.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Coagulación Sanguínea/efectos de los fármacos , Coagulantes/farmacología , Factor IXa/antagonistas & inhibidores , Factor X/antagonistas & inhibidores , Hemofilia A/diagnóstico , Tiempo de Tromboplastina Parcial , Tiempo de Protrombina , Estudios de Casos y Controles , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Humanos , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados
12.
Pharmacogenet Genomics ; 27(9): 329-336, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28678049

RESUMEN

OBJECTIVES: During anticoagulant therapy, major bleeding is one of the most severe adverse effects. This study aimed to evaluate the relationships between ABCB1, ABCG2, and CYP3A5 polymorphisms and plasma trough concentrations of apixaban, a direct inhibitor of coagulation factor X. PATIENTS AND METHODS: A total of 70 plasma concentrations of apixaban from 44 Japanese patients with atrial fibrillation were analyzed. In these analyses, the plasma trough concentration/dose (C/D) ratio of apixaban was used as a pharmacokinetic index and all data were stratified according to the presence of ABCB1 (ABCB1 1236C>T, 2677G>T/A, and 3435C>T), ABCG2 (ABCG2 421C>A), and CYP3A5 (CYP3A5*3) polymorphisms. Influences of various clinical laboratory parameters (age, serum creatinine, estimated glomerular filtration rate, aspartate amino transferase, and alanine amino transferase) on the plasma trough C/D ratio of apixaban were included in analyses. RESULTS: Although no ABCB1 polymorphisms affected the plasma trough C/D ratio of apixaban, the plasma trough C/D ratio of apixaban was significantly higher in patients with the ABCG2 421A/A genotype than in patients with the ABCG2 421C/C genotype (P<0.01). The plasma trough C/D ratio of apixaban in patients with CYP3A5*1/*3 or *3/*3 genotypes was also significantly higher than that in patients with the CYP3A5*1/*1 genotype (P<0.05). Furthermore, the plasma trough C/D ratio of apixaban decreased with increased estimated glomerular filtration rate. CONCLUSION: These results indicate that ABCG2 421A/A and CYP3A5*3 genotypes and renal function are considered potential factors affecting trough concentrations of apixaban.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Fibrilación Atrial/sangre , Citocromo P-450 CYP3A/genética , Hemorragia/genética , Proteínas de Neoplasias/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Adulto , Anciano , Anciano de 80 o más Años , Anticoagulantes/efectos adversos , Anticoagulantes/sangre , Anticoagulantes/farmacocinética , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/genética , Fibrilación Atrial/patología , Factor X/antagonistas & inhibidores , Femenino , Genotipo , Tasa de Filtración Glomerular/efectos de los fármacos , Hemorragia/inducido químicamente , Hemorragia/patología , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Pirazoles/efectos adversos , Pirazoles/sangre , Pirazoles/farmacocinética , Piridonas/efectos adversos , Piridonas/sangre , Piridonas/farmacocinética
13.
Thromb Haemost ; 117(7): 1348-1357, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28451690

RESUMEN

Emicizumab, a humanised bispecific antibody recognising factors (F) IX/IXa and X/Xa, can accelerate FIXa-catalysed FX activation by bridging FIXa and FX in a manner similar to FVIIIa. However, details of the emicizumab-antigen interactions have not been reported so far. In this study, we first showed by surface plasmon resonance analysis that emicizumab bound FIX, FIXa, FX, and FXa with moderate affinities (KD = 1.58, 1.52, 1.85, and 0.978 µM, respectively). We next showed by immunoblotting analysis that emicizumab recognised the antigens' epidermal growth factor (EGF)-like domains. We then performed KD-based simulation of equilibrium states in plasma for quantitatively predicting the ways that emicizumab would interact with the antigens. The simulation predicted that only a small part of plasma FIX, FX, and emicizumab would form antigen-bridging FIX-emicizumab-FX ternary complex, of which concentration would form a bell-shaped relationship with emicizumab concentration. The bell-shaped concentration dependency was reproduced by plasma thrombin generation assays, suggesting that the plasma concentration of the ternary complex would correlate with emicizumab's cofactor activity. The simulation also predicted that at 10.0-100 µg/ml of emicizumab-levels shown in a previous study to be clinically effective-the majority of plasma FIX, FX, and emicizumab would exist as monomers. In conclusion, emicizumab binds FIX/FIXa and FX/FXa with micromolar affinities at their EGF-like domains. The KD-based simulation predicted that the antigen-bridging ternary complex formed in circulating plasma would correlate with emicizumab's cofactor activity, and the majority of FIX and FX would be free and available for other coagulation reactions.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Factor VIIIa/inmunología , Anticuerpos Biespecíficos/sangre , Anticuerpos Monoclonales Humanizados/sangre , Especificidad de Anticuerpos , Reacciones Antígeno-Anticuerpo , Sitios de Unión , Materiales Biomiméticos/farmacología , Simulación por Computador , Factor IX/antagonistas & inhibidores , Factor IX/inmunología , Factor IXa/antagonistas & inhibidores , Factor IXa/inmunología , Factor X/antagonistas & inhibidores , Factor X/inmunología , Factor Xa/inmunología , Inhibidores del Factor Xa/sangre , Inhibidores del Factor Xa/inmunología , Inhibidores del Factor Xa/farmacología , Humanos , Modelos Inmunológicos
14.
Molecules ; 22(4)2017 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-28441767

RESUMEN

Enzymes involved in the coagulation process have received great attention as potential targets for the development of oral anti-coagulants. Among these enzymes, coagulation factor Xa (FXa) has remained the center of attention in the last decade. In this study, 16 ginsenosides and two sapogenins were isolated, identified and quantified. To determine the inhibitory potential on FXa, the chromogenic substrates method was used. The assay suggested that compounds 5, 13 and 18 were mainly responsible for the anti-coagulant effect. Furthermore, these three compounds also possessed high thrombin selectivity in the thrombin inhibition assay. Furthermore, Glide XP from Schrödinger was employed for molecular docking to clarify the interaction between the bioactive compounds and FXa. Therefore, the chemical and biological results indicate that compounds 5 (ginsenoside Rg2), 13 (ginsenoside Rg3) and 18 (protopanaxtriol, PPT) are potential natural inhibitors against FXa.


Asunto(s)
Factor X/química , Inhibidores del Factor Xa/química , Ginsenósidos/química , Panax/química , Extractos Vegetales/química , Sitios de Unión , Cromatografía Líquida de Alta Presión , Evaluación Preclínica de Medicamentos , Factor X/antagonistas & inhibidores , Inhibidores del Factor Xa/aislamiento & purificación , Inhibidores del Factor Xa/farmacología , Ginsenósidos/aislamiento & purificación , Ginsenósidos/farmacología , Cinética , Simulación del Acoplamiento Molecular , Tiempo de Tromboplastina Parcial , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Unión Proteica , Proteolisis , Trombina/antagonistas & inhibidores , Trombina/química
15.
Phlebology ; 32(6): 384-389, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27251397

RESUMEN

Postthrombotic syndrome is the most common complication after deep venous thrombosis. Postthrombotic syndrome is a debilitating disease and associated with decreased quality of life and high healthcare costs. Postthrombotic syndrome is a chronic disease, and causative treatment options are limited. Prevention of postthrombotic syndrome is therefore very important. Not all patients develop postthrombotic syndrome. Risk factors have been identified to try to predict the risk of developing postthrombotic syndrome. Age, gender, and recurrent deep venous thrombosis are factors that cannot be changed. Deep venous thrombosis location and extent seem to predict severity of postthrombotic syndrome and are potentially suitable as patient selection criteria. Residual thrombosis and reflux are known to increase the incidence of postthrombotic syndrome, but are of limited use. More recently developed treatment options for deep venous thrombosis, such as new oral factor X inhibitors and catheter-directed thrombolysis, are available at the moment. Catheter-directed thrombolysis shows promising results in reducing the incidence of postthrombotic syndrome after deep venous thrombosis. The role of new oral factor X inhibitors in preventing postthrombotic syndrome is still to be determined.


Asunto(s)
Síndrome Postrombótico/complicaciones , Síndrome Postrombótico/diagnóstico , Trombosis de la Vena/terapia , Administración Oral , Adulto , Algoritmos , Anticoagulantes/uso terapéutico , Cateterismo , Factor X/antagonistas & inhibidores , Femenino , Humanos , Incidencia , Extremidad Inferior/irrigación sanguínea , Masculino , Factores de Riesgo , Terapia Trombolítica
16.
J Thromb Haemost ; 15(3): 487-499, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28029716

RESUMEN

Essentials Capnocytophaga canimorsus causes severe dog bite related blood stream infections. We investigated if C. canimorsus contributes to bleeding abnormalities during infection. The C. canimorsus protease CcDPP7 causes factor X dysfunction by N-terminal cleavage. CcDPP7 inhibits coagulation in vivo, which could promote immune evasion and trigger hemorrhage. SUMMARY: Background Capnocytophaga canimorsus is a Gram-negative bacterium that is present in the oral flora of dogs and causes fulminant sepsis in humans who have been bitten, licked, or scratched. In patients, bleeding abnormalities, such as petechiae, purpura fulminans, or disseminated intravascular coagulation (DIC), occur frequently. Objective To investigate whether C. canimorsus could actively contribute to these bleeding abnormalities. Methods Calibrated automated thrombogram and clotting time assays were performed to assess the anticoagulant activity of C. canimorsus 5 (Cc5), a strain isolated from a fatal human infection. Clotting factor activities were measured with factor-deficient plasma. Factor X cleavage was monitored with the radiolabeled zymogen and western blotting. Mutagenesis of Cc5 genes encoding putative serine proteases was performed to identify the protease that cleaves FX. Protein purification was performed with affinity chromatography. Edman degradation allowed the detection of N-terminal cleavage of FX. Tail bleeding times were measured in mice. Results We found that Cc5 inhibited thrombin generation and increased the prothrombin time and the activated partial thromboplastin time of human plasma via FX cleavage. A mutant that was unable to synthesize a type 7 dipeptidyl peptidase (DPP7) of the S46 serine protease family failed to proteolyse FX. The purified protease (CcDPP7) cleaved FX heavy and light chains from the N-terminus, and was active in vivo after intravenous injection. Conclusions This is, to our knowledge, the first study demonstrating a detailed mechanism for FX inactivation by a bacterial protease, and it is the first functional study associating DPP7 proteases with a potentially pathogenic outcome.


Asunto(s)
Mordeduras y Picaduras/microbiología , Capnocytophaga/enzimología , Coagulación Intravascular Diseminada/microbiología , Factor X/antagonistas & inhibidores , Péptido Hidrolasas/química , Animales , Catálisis , Voluntarios Sanos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Mutación , Tiempo de Tromboplastina Parcial , Plásmidos/metabolismo , Dominios Proteicos , Sepsis/microbiología , Análisis de Secuencia de ADN
17.
Sci Rep ; 6: 32036, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27558950

RESUMEN

Unwanted clots lead to heart attack and stroke that result in a large number of deaths. Currently available anticoagulants have some drawbacks including their non-specific actions. Therefore novel anticoagulants that target specific steps in the coagulation pathway are being sought. Here we describe the identification and characterization of a novel anticoagulant protein from the venom of Hemachatus haemachatus (African Ringhals cobra) that specifically inhibits factor X (FX) activation by the extrinsic tenase complex (ETC) and thus named as exactin. Exactin belongs to the three-finger toxin (3FTx) family, with high sequence identity to neurotoxins and low identity to the well-characterized 3FTx anticoagulants-hemextin and naniproin. It is a mixed-type inhibitor of ETC with the kinetic constants, Ki' and Ki determined as 30.62 ± 7.73 nM and 153.75 ± 17.96 nM, respectively. Exactin does not bind to the active site of factor VIIa and factor Xa based on its weak inhibition (IC50 ≫ 300 µM) to the amidolytic activities of these proteases. Exactin shows exquisite macromolecular specificity to FX activation as compared to factor IX activation by ETC. Exactin thus displays a distinct mechanism when compared to other anticoagulants targeting ETC, with its selective preference to ETC-FX [ES] complex.


Asunto(s)
Anticoagulantes/farmacología , Venenos Elapídicos/química , Factor X/antagonistas & inhibidores , Hemachatus , Animales , Anticoagulantes/química , Pruebas de Coagulación Sanguínea/métodos , Dicroismo Circular , Cisteína Endopeptidasas/metabolismo , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Tiempo de Protrombina , Análisis de Secuencia de Proteína
18.
PLoS One ; 11(4): e0153770, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27089306

RESUMEN

In the present study a major protein has been purified from the venom of Indian Daboia russelii russelii using gel filtration, ion exchange and Rp-HPLC techniques. The purified protein, named daboxin P accounts for ~24% of the total protein of the crude venom and has a molecular mass of 13.597 kDa. It exhibits strong anticoagulant and phospholipase A2 activity but is devoid of any cytotoxic effect on the tested normal or cancerous cell lines. Its primary structure was deduced by N-terminal sequencing and chemical cleavage using Edman degradation and tandem mass spectrometry. It is composed of 121 amino acids with 14 cysteine residues and catalytically active His48 -Asp49 pair. The secondary structure of daboxin P constitutes 42.73% of α-helix and 12.36% of ß-sheet. It is found to be stable at acidic (pH 3.0) and neutral pH (pH 7.0) and has a Tm value of 71.59 ± 0.46°C. Daboxin P exhibits anticoagulant effect under in-vitro and in-vivo conditions. It does not inhibit the catalytic activity of the serine proteases but inhibits the activation of factor X to factor Xa by the tenase complexes both in the presence and absence of phospholipids. It also inhibits the tenase complexes when active site residue (His48) was alkylated suggesting its non-enzymatic mode of anticoagulant activity. Moreover, it also inhibits prothrombinase complex when pre-incubated with factor Xa prior to factor Va addition. Fluorescence emission spectroscopy and affinity chromatography suggest the probable interaction of daboxin P with factor X and factor Xa. Molecular docking analysis reveals the interaction of the Ca+2 binding loop; helix C; anticoagulant region and C-terminal region of daboxin P with the heavy chain of factor Xa. This is the first report of a phospholipase A2 enzyme from Indian viper venom which targets both factor X and factor Xa for its anticoagulant activity.


Asunto(s)
Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Daboia/fisiología , Factor X/antagonistas & inhibidores , Factor Xa/química , Fosfolipasas A2/farmacología , Venenos de Víboras/enzimología , Secuencia de Aminoácidos , Animales , Pruebas de Coagulación Sanguínea , Factor X/metabolismo , Factor Xa/metabolismo , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Fosfolipasas A2/química , Fosfolipasas A2/aislamiento & purificación , Estructura Secundaria de Proteína , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
19.
Arch Cardiovasc Dis ; 108(2): 122-31, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25684570

RESUMEN

BACKGROUND: Current guidelines recommend unfractionated heparin (UFH) or low-molecular-weight heparin plus an oral anticoagulant for the prevention of thromboembolism in patients undergoing electric cardioversion of atrial fibrillation (AF). Selective factor Xa inhibitors, such as fondaparinux, which has a favourable benefit-risk profile in the prevention and treatment of venous thromboembolism and the management of acute coronary syndromes, have not been systematically evaluated in this setting. AIM: To evaluate the efficacy and safety of fondaparinux versus standard treatment in patients undergoing echocardiographically-guided cardioversion of AF. METHODS: In this multicentre, randomized, open-label, controlled, two-parallel-group, phase II pilot study, patients with AF undergoing electric cardioversion following transoesophageal echocardiography (TEE) were randomized to fondaparinux or standard therapy (UFH plus vitamin K antagonist [VKA]). Patients showing an atrial thrombus in the first TEE (clot-positive) were randomized to treatment with fondaparinux or standard care for 4 weeks before cardioversion. RESULTS: The primary endpoint (combined rate of cerebral neurological events, systemic thromboembolism, all-cause death and major bleeding events) occurred in 3 of 174 (1.7%) patients on fondaparinux and 2 of 170 (1.2%) patients on UFH+VKA. The rate of thrombus disappearance among clot-positive patients was higher in the fondaparinux arm (11 of 14; 78.6%) than in the UFH+VKA arm (7 of 14; 50.0%). Incidences of adverse events were similar (45.4% with fondaparinux and 46.5% with UFH+VKA). CONCLUSION: In this pilot study in patients with TEE-guided cardioversion, the use of fondaparinux appeared to be well tolerated, with similar efficacy to UFH+VKA. Furthermore, a trend to greater thrombus resolution was observed.


Asunto(s)
Anticoagulantes/uso terapéutico , Fibrilación Atrial/terapia , Cardioversión Eléctrica , Factor X/antagonistas & inhibidores , Polisacáridos/uso terapéutico , Tromboembolia/prevención & control , Anciano , Ecocardiografía Transesofágica , Femenino , Fondaparinux , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Cirugía Asistida por Computador
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...