Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 284
Filtrar
1.
J Biol Chem ; 295(2): 517-528, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31801825

RESUMEN

Two decades of research have uncovered the mechanism by which the complex of tissue factor (TF) and the plasma serine protease factor VIIa (FVIIa) mediates the initiation of blood coagulation. Membrane-anchored TF directly interacts with substrates and induces allosteric effects in the protease domain of FVIIa. These properties are also recapitulated by the soluble ectodomain of TF (sTF). At least two interdependent allosteric activation pathways originate at the FVIIa:sTF interface are proposed to enhance FVIIa activity upon sTF binding. Here, we sought to engineer an sTF-independent FVIIa variant by stabilizing both proposed pathways, with one pathway terminating at segment 215-217 in the activation domain and the other pathway terminating at the N terminus insertion site. To stabilize segment 215-217, we replaced the flexible 170 loop of FVIIa with the more rigid 170 loop from trypsin and combined it with an L163V substitution (FVIIa-VYT). The FVIIa-VYT variant exhibited 60-fold higher amidolytic activity than FVIIa, and displayed similar FX activation and antithrombin inhibition kinetics to the FVIIa.sTF complex. The sTF-independent activity of FVIIa-VYT was partly mediated by an increase in the N terminus insertion and, as shown by X-ray crystallography, partly by Tyr-172 inserting into a cavity in the activation domain stabilizing the S1 substrate-binding pocket. The combination with L163V likely drove additional changes in a delicate hydrogen-bonding network that further stabilized S1-S3 sites. In summary, we report the first FVIIa variant that is catalytically independent of sTF and provide evidence supporting the existence of two TF-mediated allosteric activation pathways.


Asunto(s)
Coagulación Sanguínea , Factor VIIa/metabolismo , Ingeniería de Proteínas , Tromboplastina/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Cristalografía por Rayos X , Factor VIIa/química , Factor VIIa/genética , Humanos , Modelos Moleculares , Mutagénesis , Desplegamiento Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
2.
Blood ; 131(6): 674-685, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29246902

RESUMEN

The tissue factor (TF) pathway serves both hemostasis and cell signaling, but how cells control these divergent functions of TF remains incompletely understood. TF is the receptor and scaffold of coagulation proteases cleaving protease-activated receptor 2 (PAR2) that plays pivotal roles in angiogenesis and tumor development. Here we demonstrate that coagulation factor VIIa (FVIIa) elicits TF cytoplasmic domain-dependent proangiogenic cell signaling independent of the alternative PAR2 activator matriptase. We identify a Lys-Gly-Glu (KGE) integrin-binding motif in the FVIIa protease domain that is required for association of the TF-FVIIa complex with the active conformer of integrin ß1. A point mutation in this motif markedly reduces TF-FVIIa association with integrins, attenuates integrin translocation into early endosomes, and reduces delayed mitogen-activated protein kinase phosphorylation required for the induction of proangiogenic cytokines. Pharmacologic or genetic blockade of the small GTPase ADP-ribosylation factor 6 (arf6) that regulates integrin trafficking increases availability of TF-FVIIa with procoagulant activity on the cell surface, while inhibiting TF-FVIIa signaling that leads to proangiogenic cytokine expression and tumor cell migration. These experiments delineate the structural basis for the crosstalk of the TF-FVIIa complex with integrin trafficking and suggest a crucial role for endosomal PAR2 signaling in pathways of tissue repair and tumor biology.


Asunto(s)
Factor VIIa/química , Factor VIIa/metabolismo , Integrina beta1/metabolismo , Dominios y Motivos de Interacción de Proteínas , Receptor PAR-2/metabolismo , Factor 6 de Ribosilación del ADP , Animales , Sitios de Unión/genética , Células Cultivadas , Factor VIIa/genética , Humanos , Integrina beta1/química , Ratones , Células 3T3 NIH , Neovascularización Fisiológica/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Mapas de Interacción de Proteínas , Receptor PAR-2/genética , Transducción de Señal/genética , Tromboplastina/química , Tromboplastina/metabolismo
3.
Proc Natl Acad Sci U S A ; 114(47): 12454-12459, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109275

RESUMEN

Recombinant factor VIIa (FVIIa) variants with increased activity offer the promise to improve the treatment of bleeding episodes in patients with inhibitor-complicated hemophilia. Here, an approach was adopted to enhance the activity of FVIIa by selectively optimizing substrate turnover at the membrane surface. Under physiological conditions, endogenous FVIIa engages its cell-localized cofactor tissue factor (TF), which stimulates activity through membrane-dependent substrate recognition and allosteric effects. To exploit these properties of TF, a covalent complex between FVIIa and the soluble ectodomain of TF (sTF) was engineered by introduction of a nonperturbing cystine bridge (FVIIa Q64C-sTF G109C) in the interface. Upon coexpression, FVIIa Q64C and sTF G109C spontaneously assembled into a covalent complex with functional properties similar to the noncovalent wild-type complex. Additional introduction of a FVIIa-M306D mutation to uncouple the sTF-mediated allosteric stimulation of FVIIa provided a final complex with FVIIa-like activity in solution, while exhibiting a two to three orders-of-magnitude increase in activity relative to FVIIa upon exposure to a procoagulant membrane. In a mouse model of hemophilia A, the complex normalized hemostasis upon vascular injury at a dose of 0.3 nmol/kg compared with 300 nmol/kg for FVIIa.


Asunto(s)
Terapia Biológica/métodos , Factor VIIa/química , Hemofilia A/terapia , Ingeniería de Proteínas/métodos , Tromboplastina/química , Regulación Alostérica , Animales , Coagulación Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Factor VIIa/genética , Factor VIIa/farmacología , Factor VIIa/uso terapéutico , Femenino , Hemofilia A/fisiopatología , Humanos , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Simulación de Dinámica Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Tromboplastina/genética , Tromboplastina/farmacología , Tromboplastina/uso terapéutico
4.
Biophys J ; 116(10): 1823-1835, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31003762

RESUMEN

A critical step in injury-induced initiation of blood coagulation is the formation of the complex between the trypsin-like protease coagulation factor VIIa (FVIIa) and its cofactor tissue factor (TF), which converts FVIIa from an intrinsically poor enzyme to an active protease capable of activating zymogens of downstream coagulation proteases. Unlike its constitutively active ancestor trypsin, FVIIa is allosterically activated (by TF). Here, ensemble refinement of crystallographic structures, which uses multiple copies of the entire structure as a means of representing structural flexibility, is applied to explore the impacts of inhibitor binding to trypsin and FVIIa, as well as cofactor binding to FVIIa. To assess the conformational flexibility and its role in allosteric pathways in these proteases, main-chain hydrogen bond networks are analyzed by calculating the hydrogen-bond propensity. Mapping pairwise propensity differences between relevant structures shows that binding of the inhibitor benzamidine to trypsin has a minor influence on the protease flexibility. For FVIIa, in contrast, the protease domain is "locked" into the catalytically competent trypsin-like configuration upon benzamidine binding as indicated by the stabilization of key structural features: the nonprime binding cleft and the oxyanion hole are stabilized, and the effect propagates from the active site region to the calcium-binding site and to the vicinity of the disulphide bridge connecting with the light chain. TF binding to FVIIa furthermore results in stabilization of the 170 loop, which in turn propagates an allosteric signal from the TF-binding region to the active site. Analyses of disulphide bridge energy and flexibility reflect the striking stability difference between the unregulated enzyme and the allosterically activated form after inhibitor or cofactor binding. The ensemble refinement analyses show directly, for the first time to our knowledge, whole-domain structural footprints of TF-induced allosteric networks present in x-ray crystallographic structures of FVIIa, which previously only have been hypothesized or indirectly inferred.


Asunto(s)
Factor VIIa/química , Factor VIIa/metabolismo , Regulación Alostérica , Apoenzimas/química , Apoenzimas/metabolismo , Benzamidinas/farmacología , Cristalografía por Rayos X , Disulfuros/química , Activación Enzimática/efectos de los fármacos , Modelos Moleculares , Dominios Proteicos , Pliegue de Proteína , Tripsina/química , Tripsina/metabolismo , Tripsinógeno/metabolismo
5.
J Biol Chem ; 292(39): 16249-16256, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28801460

RESUMEN

Interactions of soluble proteins with the cell membrane are critical within the blood coagulation cascade. Of particular interest are the interactions of γ-carboxyglutamic acid-rich domain-containing clotting proteins with lipids. Variability among conventional analytical methods presents challenges for comparing clotting protein-lipid interactions. Most previous studies have investigated only a single clotting protein and lipid composition and have yielded widely different binding constants. Herein, we demonstrate that a combination of lipid bilayer nanodiscs and a multiplexed silicon photonic analysis technology enables high-throughput probing of many protein-lipid interactions among blood-clotting proteins. This approach allowed direct comparison of the binding constants of prothrombin, factor X, activated factor VII, and activated protein C to seven different binary lipid compositions. In a single experiment, the binding constants of one protein interacting with all lipid compositions were simultaneously determined. A simple surface regeneration then facilitated similar binding measurements for three other coagulation proteins. As expected, our results indicated that all proteins exhibit tighter binding (lower Kd ) as the proportion of anionic lipid increases. Interestingly, at high proportions of phosphatidylserine, the Kd values of all four proteins began to converge. We also found that although koff values for all four proteins followed trends similar to those observed for the Kd values, the variation among the proteins was much lower, indicating that much of the variation came from the kinetic binding (kon) of the proteins. These findings indicate that the combination of silicon photonic microring resonator arrays and nanodiscs enables rapid interrogation of biomolecular binding interactions at model cell membrane interfaces.


Asunto(s)
Factor VIIa/metabolismo , Factor X/metabolismo , Ácidos Fosfatidicos/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidilserinas/metabolismo , Proteína C/metabolismo , Protrombina/metabolismo , Factor VIIa/química , Factor VIIa/genética , Factor X/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Cinética , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Nanoestructuras/química , Fenómenos Ópticos , Ácidos Fosfatidicos/química , Fosfatidilcolinas/química , Fosfatidilserinas/química , Análisis por Matrices de Proteínas , Proteína C/química , Protrombina/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Silicio/química
6.
Haemophilia ; 24(3): 477-486, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29537116

RESUMEN

INTRODUCTION: Recombinant FVIIa (rFVIIa) is an effective treatment for haemophilia through frequent administration. However, the short half-life of rFVIIa decreases its prophylactic ability to reduce bleeding. Carboxy-terminal peptide (CTP)-modified FVIIa (MOD-5014) is a long-acting rFVIIa developed for on-demand treatment of haemophilia using either an intravenous or subcutaneous injection with the aim of less frequent administrations, as well as for prophylactic use. AIM: The comprehensive evaluation of the activity MOD-5014 vs commercially available rhFVIIa, as well as their interaction with cofactors and inhibitors. METHODS: The in vitro characterization included clotting activity, affinity by surface plasmon resonance, cleavage of synthetic substrates, thrombin generation (TG) and rotation thromboelastometry. RESULTS: Reduced specific activity was obtained for MOD-5014 compared to rhFVIIa, while both compounds demonstrated comparable affinity to tissue factor (TF). MOD-5014 showed reduced TG when spiked at a similar concentration as rhFVIIa, suggesting that an increased concentration might be needed in a clinical setting to provide initial haemostatic effect. MOD-5014 demonstrated a slightly lower affinity for binding to activated platelets and slightly lower proteolytic activity on the platelet surface, possibly as the fusion of CTP has the potential to sterically hinder binding to both the platelet membrane and to protein substrates. Both compounds showed a similar dose-dependent stimulatory effect on clot formation, and both showed a similar deactivation pattern following incubation with TF pathway inhibitor (TFPI), antithrombin and heparin. CONCLUSION: The comparable in vitro activity of MOD-5014 and rhFVIIa paves the way for in vivo pharmacology evaluations of MOD-5014 in preparation for clinical studies.


Asunto(s)
Factor VIIa/química , Factor VIIa/farmacología , Coagulación Sanguínea/efectos de los fármacos , Factor VIIa/administración & dosificación , Factor VIIa/metabolismo , Humanos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Tromboplastina/metabolismo
7.
J Biol Chem ; 291(9): 4671-83, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26694616

RESUMEN

The complex of coagulation factor VIIa (FVIIa), a trypsin-like serine protease, and membrane-bound tissue factor (TF) initiates blood coagulation upon vascular injury. Binding of TF to FVIIa promotes allosteric conformational changes in the FVIIa protease domain and improves its catalytic properties. Extensive studies have revealed two putative pathways for this allosteric communication. Here we provide further details of this allosteric communication by investigating FVIIa loop swap variants containing the 170 loop of trypsin that display TF-independent enhanced activity. Using x-ray crystallography, we show that the introduced 170 loop from trypsin directly interacts with the FVIIa active site, stabilizing segment 215-217 and activation loop 3, leading to enhanced activity. Molecular dynamics simulations and novel fluorescence quenching studies support that segment 215-217 conformation is pivotal to the enhanced activity of the FVIIa variants. We speculate that the allosteric regulation of FVIIa activity by TF binding follows a similar path in conjunction with protease domain N terminus insertion, suggesting a more complete molecular basis of TF-mediated allosteric enhancement of FVIIa activity.


Asunto(s)
Factor VIIa/metabolismo , Modelos Moleculares , Tromboplastina/metabolismo , Tripsina/metabolismo , Regulación Alostérica , Dominio Catalítico , Cristalografía por Rayos X , Estabilidad de Enzimas , Factor VIIa/química , Factor VIIa/genética , Humanos , Cinética , Simulación de Dinámica Molecular , Mutación , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Desplegamiento Proteico , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Solubilidad , Tromboplastina/química , Tromboplastina/genética , Tripsina/química , Tripsina/genética
8.
Haemophilia ; 23(4): e324-e334, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28594467

RESUMEN

BACKGROUND: The bypassing agent factor VII (FVIIa) is a first-line therapy for the treatment of acute bleeding episodes in patients with haemophilia and high-titre inhibitors. FVIIa is a highly post-translationally modified protein that requires eukaryotic expression systems to produce a fully active molecule. A recombinant FVIIa was produced in the milk of transgenic rabbits to increase expression and provide an efficient, safe and affordable product after purification to homogeneity (LR769). AIM: To present the biochemical and functional in vitro characteristics of LR769. RESULTS: Mass spectrometric analyses of the intact protein and of heavy and light chains revealed a fully activated, mature and properly post-translationally modified protein notably regarding N/O-glycosylations and γ-carboxylation. Primary structure analysis, performed by peptide mapping, confirmed 100% of the sequence and the low level or absence of product-derived impurities such as oxidized, deamidated and glycated forms. Low levels of aggregates and fragments were observed by different chromatographic methods. Higher order structure investigated by circular dichroism showed appropriate secondary/tertiary structures and conformational change in the presence of Ca2+ ions. Finally, activated partial thromboplastin time and thrombin generation assays showed the ability of LR769 to decrease coagulation time and to generate thrombin in haemophiliac-A-plasmas, even in the presence of inhibitors. CONCLUSION: The innovative expression system used to produce LR769 yields a new safe and effective rhFVIIa for the treatment of haemophilia A or B patients with inhibitors.


Asunto(s)
Factor VIIa/química , Factor VIIa/metabolismo , Leche/metabolismo , Animales , Animales Modificados Genéticamente , Factor VIIa/biosíntesis , Factor VIIa/genética , Humanos , Tiempo de Tromboplastina Parcial , Conejos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombina/biosíntesis
9.
J Chem Inf Model ; 57(7): 1652-1666, 2017 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-28565907

RESUMEN

Water molecules play an essential role for mediating interactions between ligands and protein binding sites. Displacement of specific water molecules can favorably modulate the free energy of binding of protein-ligand complexes. Here, the nature of water interactions in protein binding sites is investigated by 3D RISM (three-dimensional reference interaction site model) integral equation theory to understand and exploit local thermodynamic features of water molecules by ranking their possible displacement in structure-based design. Unlike molecular dynamics-based approaches, 3D RISM theory allows for fast and noise-free calculations using the same detailed level of solute-solvent interaction description. Here we correlate molecular water entities instead of mere site density maxima with local contributions to the solvation free energy using novel algorithms. Distinct water molecules and hydration sites are investigated in multiple protein-ligand X-ray structures, namely streptavidin, factor Xa, and factor VIIa, based on 3D RISM-derived free energy density fields. Our approach allows the semiquantitative assessment of whether a given structural water molecule can potentially be targeted for replacement in structure-based design. Finally, PLS-based regression models from free energy density fields used within a 3D-QSAR approach (CARMa - comparative analysis of 3D RISM Maps) are shown to be able to extract relevant information for the interpretation of structure-activity relationship (SAR) trends, as demonstrated for a series of serine protease inhibitors.


Asunto(s)
Simulación de Dinámica Molecular , Proteínas/química , Proteínas/metabolismo , Sitios de Unión , Proteínas Sanguíneas/química , Proteínas Sanguíneas/farmacología , Clorobenzoatos/química , Clorobenzoatos/farmacología , Factor VIIa/química , Factor VIIa/metabolismo , Factor Xa/química , Factor Xa/metabolismo , Inhibidores del Factor Xa/química , Inhibidores del Factor Xa/farmacología , Ligandos , Unión Proteica , Conformación Proteica , Proteínas/antagonistas & inhibidores , Relación Estructura-Actividad Cuantitativa , Estreptavidina/química , Estreptavidina/metabolismo , Termodinámica , Agua/metabolismo
10.
Phys Chem Chem Phys ; 19(33): 22230-22242, 2017 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-28799584

RESUMEN

During blood-coagulation, the transmembrane protein tissue factor (TF) binds to its ligand, factor (F)VII, activating and allosterically modifying it to form a mature active binary complex (TF-FVIIa). Although the extracellular domain of TF (sTF) can bind to FVII, it fails to activate it. Binding of TF with FVIIa only partially enhances FVIIa proteolytic activity. Our previous kinetic study revealed that sTF has a lower binding capacity with FVIIa compared to membrane bound full-length (fl)TF. The reason behind this incapability of FVII activation and reduced catalytic activity remains unexplored due to the lack of an flTF crystal structure. Here we employed a comparative dynamic study between sTF-FVIIa in solution and flTF-FVIIa in a membrane system to give probable explanations for the differential behaviour of these complexes. Based on potential of mean force and interaction energy calculations, the binding affinities between sTF and FVIIa are weaker than those of the flTF-FVIIa complex. We further observed domain-wise less stability, reduced height, and thus less inter and intra-domain interaction between the sTF and FVIIa complexes. We detected higher fluctuation among the inter-atomic distances of the catalytic triad (CT) residues in sTF-FVIIa over the flTF-FVIIa complex. The flTF-FVIIa complex forms two major interactions between EGF2 and TF. We showed the enhanced activity of the flTF-FVIIa complex over the sTF-FVIIa complex, which is guided by mainly two interactions between EGF2 and TF. Due to the lack of these interactions, sTF-FVIIa somehow forms a less stable binary complex and could not react upon binding its substrates (FIX, FX). Our study, for the first time, provides a possible explanation of the distinct behaviour of the two forms of TF (sTF and flTF) towards its only ligand FVII/FVIIa.


Asunto(s)
Factor VIIa/metabolismo , Tromboplastina/metabolismo , Regulación Alostérica , Sitios de Unión , Biocatálisis , Activación Enzimática , Factor VIIa/química , Humanos , Cinética , Ligandos , Simulación de Dinámica Molecular , Unión Proteica , Dominios Proteicos , Estructura Terciaria de Proteína , Termodinámica , Tromboplastina/química
11.
Biochemistry ; 54(30): 4665-71, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26169722

RESUMEN

The blood coagulation cascade is initiated when the cell-surface complex of factor VIIa (FVIIa, a trypsin-like serine protease) and tissue factor (TF, an integral membrane protein) proteolytically activates factor X (FX). Both FVIIa and FX bind to membranes via their γ-carboxyglutamate-rich domains (GLA domains). GLA domains contain seven to nine bound Ca(2+) ions that are critical for their folding and function, and most biochemical studies of blood clotting have employed supraphysiologic Ca(2+) concentrations to ensure saturation of these domains with bound Ca(2+). Recently, it has become clear that, at plasma concentrations of metal ions, Mg(2+) actually occupies two or three of the divalent metal ion-binding sites in GLA domains, and that these bound Mg(2+) ions are required for full function of these clotting proteins. In this study, we investigated how Mg(2+) influences FVIIa enzymatic activity. We found that the presence of TF was required for Mg(2+) to enhance the rate of FX activation by FVIIa, and we used alanine-scanning mutagenesis to identify TF residues important for mediating this response to Mg(2+). Several TF mutations, including those at residues G164, K166, and Y185, blunted the ability of Mg(2+) to enhance the activity of the TF/FVIIa complex. Our results suggest that these TF residues interact with the GLA domain of FX in a Mg(2+)-dependent manner (although effects of Mg(2+) on the FVIIa GLA domain cannot be ruled out). Notably, these TF residues are located within or immediately adjacent to the putative substrate-binding exosite of TF.


Asunto(s)
Factor VIIa/química , Magnesio/química , Complejos Multiproteicos/química , Tromboplastina/química , Sustitución de Aminoácidos , Calcio/química , Calcio/metabolismo , Factor VIIa/genética , Factor VIIa/metabolismo , Factor X/química , Factor X/genética , Factor X/metabolismo , Humanos , Magnesio/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutación Missense , Estructura Terciaria de Proteína , Tromboplastina/genética , Tromboplastina/metabolismo
12.
J Biol Chem ; 289(51): 35388-96, 2014 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-25344622

RESUMEN

Factor VIIa (FVIIa) is a trypsin-like protease that plays an important role in initiating blood coagulation. Very limited structural information is available for the free, inactive form of FVIIa that circulates in the blood prior to vascular injury and the molecular details of its activity enhancement remain elusive. Here we have applied hydrogen/deuterium exchange mass spectrometry coupled to electron transfer dissociation to pinpoint individual residues in the heavy chain of FVIIa whose conformation and/or local interaction pattern changes when the enzyme transitions to the active form, as induced either by its cofactor tissue factor or a covalent active site inhibitor. Identified regulatory residues are situated at key sites across one continuous surface of the protease domain spanning the TF-binding helix across the activation pocket to the calcium binding site and are embedded in elements of secondary structure and at the base of flexible loops. Thus these residues are optimally positioned to mediate crosstalk between functional sites in FVIIa, particularly the cofactor binding site and the active site. Our results unambiguously show that the conformational allosteric activation signal extends to the EGF1 domain in the light chain of FVIIa, underscoring a remarkable intra- and interdomain allosteric regulation of this trypsin-like protease.


Asunto(s)
Medición de Intercambio de Deuterio/métodos , Factor VIIa/química , Espectrometría de Masas/métodos , Estructura Terciaria de Proteína , Sitio Alostérico , Secuencia de Aminoácidos , Sitios de Unión , Calcio/química , Calcio/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Transporte de Electrón , Factor VIIa/metabolismo , Glicosilación , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Tromboplastina/química , Tromboplastina/metabolismo
13.
J Biol Chem ; 289(3): 1732-41, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24275667

RESUMEN

Tissue factor pathway inhibitor (TFPI) is a Kunitz-type protease inhibitor that inhibits activated factor X (FXa) via a slow-tight binding mechanism and tissue factor-activated FVII (TF-FVIIa) via formation of a quaternary FXa-TFPI-TF-FVIIa complex. Inhibition of TFPI enhances coagulation in hemophilia models. Using a library approach, we selected and subsequently optimized peptides that bind TFPI and block its anticoagulant activity. One peptide (termed compound 3), bound with high affinity to the Kunitz-1 (K1) domain of TFPI (Kd ∼1 nM). We solved the crystal structure of this peptide in complex with the K1 of TFPI at 2.55-Å resolution. The structure of compound 3 can be segmented into a N-terminal anchor; an Ω-shaped loop; an intermediate segment; a tight glycine-loop; and a C-terminal α-helix that is anchored to K1 at its reactive center loop and two-stranded ß-sheet. The contact surface has an overall hydrophobic character with some charged hot spots. In a model system, compound 3 blocked FXa inhibition by TFPI (EC50 = 11 nM) and inhibition of TF-FVIIa-catalyzed FX activation by TFPI (EC50 = 2 nM). The peptide prevented transition from the loose to the tight FXa-TFPI complex, but did not affect formation of the loose FXa-TFPI complex. The K1 domain of TFPI binds and inhibits FVIIa and the K2 domain similarly inhibits FXa. Because compound 3 binds to K1, our data show that K1 is not only important for FVIIa inhibition but also for FXa inhibition, i.e. for the transition of the loose to the tight FXa-TFPI complex. This mode of action translates into normalization of coagulation of hemophilia plasmas. Compound 3 thus bears potential to prevent bleeding in hemophilia patients.


Asunto(s)
Coagulantes/química , Factor VIIa/química , Factor Xa/química , Lipoproteínas/antagonistas & inhibidores , Péptidos/química , Coagulación Sanguínea/efectos de los fármacos , Coagulantes/síntesis química , Coagulantes/metabolismo , Coagulantes/uso terapéutico , Factor VIIa/metabolismo , Factor Xa/metabolismo , Hemofilia A/tratamiento farmacológico , Hemofilia A/metabolismo , Hemorragia/tratamiento farmacológico , Hemorragia/metabolismo , Humanos , Lipoproteínas/química , Lipoproteínas/metabolismo , Péptidos/síntesis química , Péptidos/metabolismo , Péptidos/uso terapéutico , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
14.
BMC Biotechnol ; 15: 87, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26382581

RESUMEN

UNLABELLED: BACKGROUND & METHODS: Recombinant factor VII (rFVII), the precursor molecule for recombinant activated FVII (rFVIIa), is, due to its need for complex post translational modifications, produced in mammalian cells. To evaluate the suitability of a human cell line in order to produce rFVII with post-translational modifications as close as possible to pdFVII, we compared the biochemical properties of rFVII synthesized in human embryonic kidney-derived (HEK)293 cells (HEK293rFVII) with those of rFVII expressed in Chinese hamster ovary (CHO, CHOrFVII) and baby hamster kidney (BHK, BHKrFVII) cells, and also with those of plasma derived FVII (pdFVII), using various analytical methods. rFVII was purified from selected production clones derived from BHK, CHO, and HEK293 cells after stable transfection, and rFVII isolates were analyzed for protein activity, impurities and post-translational modifications. RESULTS & DISCUSSION: The analytical results showed no apparent gross differences between the various FVII proteins, except in their N-linked glycosylation pattern. Most N-glycans found on rFVII produced in HEK293 cells were not detected on rFVII from CHO and BHK cells, or, somewhat unexpectedly, on pdFVII; all other protein features were similar. HEK293rFVII glycans were mainly characterized by a higher structural variety and a lower degree of terminal sialylation, and a high amount of terminal N-acetyl galactosamines (GalNAc). All HEK293rFVII oligosaccharides contained one or more fucoses (Fuc), as well as hybrid and high mannose (Man) structures. CONCLUSIONS: From all rFVII isolates investigated, CHOrFVII contained the highest degree of sialylation and no terminal GalNAc, and CHO cells were therefore assumed to be the best option for the production of rFVII.


Asunto(s)
Factor VIIa/química , Factor VIIa/metabolismo , Glicosilación , Animales , Células CHO , Secuencia de Carbohidratos , Cricetinae , Cricetulus , Células HEK293 , Humanos , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
15.
Semin Thromb Hemost ; 41(7): 682-90, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26408924

RESUMEN

Interactions between tissue factor and factor VIIa are the primary initiators of coagulation in hemostasis and certain thrombotic diseases. Tissue factor, an integral membrane protein expressed extensively outside of the vasculature, is the regulatory protein cofactor for coagulation factor VIIa. Factor VIIa, a trypsin-like serine protease homologous with other blood coagulation proteases, is weakly active when free in solution and must bind its membrane-bound cofactor for physiologically relevant activity. Tissue factor allosterically activates factor VIIa by several mechanisms such as active site positioning, spatial stabilization, and direct interactions with the substrate. Protein-membrane interactions between tissue factor, factor VIIa, and substrates all play critical roles in modulating the activity of this enzyme complex. Additionally, divalent cations such as Ca(2+) and Mg(2+) are critical for correct protein folding, as well as protein-membrane and protein-protein interactions. The contributions of these factors toward tissue factor-factor VIIa activity are discussed in this review.


Asunto(s)
Factor VIIa/química , Factor VIIa/metabolismo , Pliegue de Proteína , Tromboplastina/química , Tromboplastina/metabolismo , Regulación Alostérica , Animales , Calcio/química , Calcio/metabolismo , Humanos , Magnesio/química , Magnesio/metabolismo , Relación Estructura-Actividad
16.
Biochim Biophys Acta ; 1830(6): 3489-96, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23434438

RESUMEN

BACKGROUND: Tissue factor (TF), an in vivo initiator of blood coagulation, is a transmembrane protein and has two disulfides in the extracellular domain. The integrity of one cysteine pair, Cys186-Cys209, has been hypothesized to be essential for an allosteric "decryption" phenomenon, presumably regulating TF procoagulant function, which has been the subject of a lengthy debate. The conclusions of published studies on this subject are based on indirect evidences obtained by the use of reagents with potentially oxidizing/reducing properties. METHODS: The status of disulfides in recombinant TF1-263 and natural placental TF in their non-reduced native and reduced forms was determined by mass-spectrometry. Functional assays were performed to assess TF cofactor function. RESULTS: In native proteins, all four cysteines of the extracellular domain of TF are oxidized. Reduced TF retains factor VIIa binding capacity but completely loses the cofactor function. CONCLUSION: The reduction of TF disulfides (with or without alkylation) eliminates TF regulation of factor VIIa catalytic function in both membrane dependent FX activation and membrane independent synthetic substrate hydrolysis. GENERAL SIGNIFICANCE: Results of this study advance our knowledge on TF structure/function relationships.


Asunto(s)
Apoenzimas/química , Disulfuros/química , Tromboplastina/química , Regulación Alostérica/fisiología , Apoenzimas/metabolismo , Coagulación Sanguínea/fisiología , Coenzimas/química , Coenzimas/metabolismo , Disulfuros/metabolismo , Factor VIIa/química , Factor VIIa/metabolismo , Factor X/química , Factor X/metabolismo , Humanos , Oxidación-Reducción , Proteínas Gestacionales/química , Proteínas Gestacionales/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Tromboplastina/metabolismo
17.
Transfus Apher Sci ; 50(1): 20-5, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24529682

RESUMEN

Bleeding related to cardiac surgery is an important clinical problem. Perioperative anemia and transfusion of allogeneic blood products have both been associated with adverse outcome including mortality and major morbidity. Guidelines exist to help determine when the risks of anemia outweigh the risks of transfusion. Perioperative bleeding may be related to several factors including the use of new antithrombotic drugs. A variety of hemostatic drugs have been studied to reduce bleeding and transfusion, although several questions and concerns about them exist. Patient blood management programs can be valuable for management of patients undergoing cardiac surgery.


Asunto(s)
Pérdida de Sangre Quirúrgica/prevención & control , Procedimientos Quirúrgicos Cardíacos/métodos , Reacción a la Transfusión , Anemia/etiología , Anticoagulantes/uso terapéutico , Conservación de la Sangre , Factor VIIa/química , Fibrinógeno/química , Fibrinolíticos/uso terapéutico , Hemorragia/inducido químicamente , Hemostasis , Humanos , Periodo Perioperatorio , Proteínas Recombinantes/química , Riesgo
18.
J Biol Chem ; 287(12): 8994-9001, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22275370

RESUMEN

In the absence of its cofactor tissue factor (TF), coagulation factor VIIa (FVIIa) predominantly exists in a zymogen-like, catalytically incompetent state. Here we demonstrate that conformation-specific monoclonal antibodies (mAbs) can be used to characterize structural features determining the activity of FVIIa. We isolated two classes of mAbs, which both increased the catalytic efficiency of FVIIa more than 150-fold. The effects of the antibodies were retained with a FVIIa variant, which has been shown to be inert to allosteric activation by the natural activator TF, suggesting that the antibodies and TF employ distinct mechanisms of activation. The antibodies could be classified into two groups based on their patterns of affinities for different conformations of FVIIa. Whereas one class of antibodies affected both the K(m) and k(cat), the other class mainly affected the K(m). The antibody-induced activity enhancement could be traced to maturation of the S1 substrate binding pocket and the oxyanion hole, evident by an increased affinity for p-aminobenzamidine, an increased rate of antithrombin inhibition, an increased rate of incorporation of diisopropylfluorophosphate, and an enhanced fraction of molecules with a buried N terminus of the catalytic domain in the presence of antibodies. As demonstrated by site-directed mutagenesis, the two groups of antibodies appear to have overlapping, although clearly different, epitopes in the 170-loop. Our findings suggest that binding of ligands to specific residues in the 170-loop or its spatial vicinity may stabilize the S1 pocket and the oxyanion hole, and they may have general implications for the molecular understanding of FVIIa regulatory mechanisms.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Factor VIIa/química , Factor VIIa/metabolismo , Regulación Alostérica , Animales , Anticuerpos Monoclonales/química , Sitios de Unión , Factor VIIa/genética , Humanos , Cinética , Ratones , Unión Proteica , Tromboplastina/genética , Tromboplastina/metabolismo
19.
Glycobiology ; 23(12): 1531-46, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24092837

RESUMEN

Human coagulation factor VIIa is a glycoprotein that promotes haemostasis through activation of the coagulation cascade extrinsic pathway. Most haemophilia A/B patients with inhibitors are treated by injection of plasma-derived or recombinant FVIIa. The use of recombinant products raises questions about the ability of the host cell to produce efficiently post-translationally modified proteins. Glycosylation is especially critical considering that it can modulate protein safety and efficacy. The present paper reports the N-/O-glycosylation pattern of a new recombinant human factor VIIa expressed in the mammary glands of transgenic rabbits. Glycosylation was investigated by chromatography and advanced mass spectrometry techniques for glycan identification and quantitation. Mass spectrometry (MS)/MS analyses were performed to confirm the glycan structures as well as the position and branching of specific monosaccharides or substituents. The two N-glycosylation sites were found to be fully occupied mostly by mono- and bi-sialylated biantennary complex-type structures, the major form being A(2)G(2)S(1). Some oligomannose/hybrid structures were retrieved in lower abundance, the major ones being GlcNAcα1,O-phosphorylated at the C6-position of a Man residue (Man-6-(GlcNAcα1,O-)phosphate motif) as commonly observed on lysosomal proteins. No immunogenic glycotopes such as Galili (Galα1,3Gal) and HD antigens (N-glycolylneuraminic acid (NeuGc)) were detected. Concerning O-glycosylation, the product exhibited O-fucose and O-glucose-(xylose)(0, 1, 2) motifs as expected. The N-glycosylation consistency was also investigated by varying production parameters such as the period of lactation, the number of consecutive lactations and rabbit generations. Results show that the transgenesis technology is suitable for the long-term production of rhFVIIa with a reproducible glycosylation pattern.


Asunto(s)
Factor VIIa/biosíntesis , Factor VIIa/química , Leche/química , Leche/metabolismo , Animales , Animales Modificados Genéticamente , Factor VIIa/análisis , Factor VIIa/genética , Glicosilación , Humanos , Glándulas Mamarias Humanas/metabolismo , Conejos
20.
Blood ; 117(15): 3974-82, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21325603

RESUMEN

Catalytic domain variants of activated factor VII (FVIIa) with enhanced hemostatic properties are highly attractive for the treatment of bleeding disorders via gene-based therapy. To explore this in a hemophilic mouse model, we characterized 2 variants of murine activated FVII (mFVIIa-VEAY and mFVIIa-DVQ) with modified catalytic domains, based on recombinant human FVIIa (rhFVIIa) variants. Using purified recombinant proteins, we showed that murine FVIIa (mFVIIa) and variants had comparable binding to human and murine tissue factor (TF) and exhibited similar extrinsic coagulant activity. In vitro in the absence of TF, the variants showed a 6- to 17-fold enhanced proteolytic and coagulant activity relative to mFVIIa, but increased inactivation by antithrombin. Gene delivery of mFVIIa-VEAY resulted in long-term, effective hemostasis at 5-fold lower expression levels relative to mFVIIa in hemophilia A mice or in hemophilia B mice with inhibitors to factor IX. However, expression of mFVIIa-VEAY at 14-fold higher than therapeutic levels resulted in a progressive mortality to 70% within 6 weeks after gene delivery. These results are the first demonstration of the hemostatic efficacy of continuous expression, in the presence or absence of inhibitors, of a high-activity gene-based FVIIa variant in an animal model of hemophilia.


Asunto(s)
Dominio Catalítico/genética , Factor VIIa/genética , Terapia Genética/métodos , Hemofilia A/terapia , Hemostasis/fisiología , Animales , Línea Celular , Dependovirus/genética , Modelos Animales de Enfermedad , Factor VIIa/química , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/mortalidad , Hemofilia A/sangre , Hemofilia A/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Plásmidos/genética , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA