Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 295(2): 517-528, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31801825

ABSTRACT

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.


Subject(s)
Blood Coagulation , Factor VIIa/metabolism , Protein Engineering , Thromboplastin/metabolism , Allosteric Regulation , Amino Acid Sequence , Crystallography, X-Ray , Factor VIIa/chemistry , Factor VIIa/genetics , Humans , Models, Molecular , Mutagenesis , Protein Unfolding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
2.
Res Pract Thromb Haemost ; 3(4): 713-717, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31624791

ABSTRACT

BACKGROUND: Thrombin generation assay (TGA) and thrombelastography (TEG) are increasingly employed, global, in vitro methods for assessment of the procoagulant potential of plasma/blood and possibly ideally suited tools to monitor, for example, therapy with recombinant factor VIIa (FVIIa). It remains controversial to what extent results obtained with spiked and postinfusion samples reflect the outcome in patients. OBJECTIVE: To characterize the TGA response to FVIIa in hemophilic plasma and compare with TEG data. METHODS: Hemophilia A (HA) was induced in platelet-rich plasma (PRP) from healthy volunteers, followed by spiking with FVIIa, γ-carboxyglutamic acid (Gla)-domainless FVIIa or V158D/E296V/M298Q-FVIIa (FVIIaDVQ). Samples were triggered with tissue factor and analyzed by TGA and TEG in parallel. RESULTS: Addition of 25 nmol L-1 FVIIa to HA PRP normalized TEG parameters angle and R time, as well as TGA lag time, but had poor effects on the thrombin peak height and velocity index. All parameters (at least) returned to normal levels either upon adding a much higher concentration of FVIIa (~1500 nmol L-1) or by using the superactive variant FVIIaDVQ. Surprisingly, Gla-domainless derivatives of FVIIa and FVIIaDVQ also yielded considerable effects in HA PRP. CONCLUSIONS: The good general responses to clinically effective concentrations of FVIIa (25 and 75 nmol L-1) seen in TEG analyses, as well as for TGA lag time, were accompanied by far-from-normal thrombin peaks. A near-normal thrombin peak response required the presence of considerably higher FVIIa activity but, intriguingly, relied only marginally on a functional Gla domain (ie, on platelet surface localization).

3.
Res Pract Thromb Haemost ; 3(3): 412-419, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31294329

ABSTRACT

BACKGROUND: Blood coagulation factor VIIa (FVIIa) plays its critical physiological role in the initiation of hemostasis. Even so, recombinant FVIIa is successfully used as a bypassing agent for factor VIII or IX in the treatment of bleeds in patients with severe hemophilia with inhibitors. To investigate the utility of more potent FVIIa variants with enhanced intrinsic activity, molecules such as V21D/E154V/M156Q-FVIIa (FVIIaDVQ) were designed. METHODS: Surface plasmon resonance was used to characterize the binding of mAb4F5 to FVIIaDVQ and related variants. X-ray crystallography was used to determine the structure of the Fab fragment of mAb4F5 (Fab4F5). Molecular docking and small angle X-ray scattering led to a model of FVIIaDVQ:Fab4F5 complex. RESULTS: The binding experiments, functional effects on FVIIaDVQ and structure of mAb4F5 (originally intended for quantification of FVIIaDVQ in samples containing FVII(a)) pinpointed the epitope (crucial role for residue Asp21) and shed light on the role of the N-terminus of the protease domain in FVIIa allostery. The potential antigen-combining sites are composed of 1 hydrophobic and 1 negatively charged pocket formed by 6 complementarity-determining region (CDR) loops. Structural analysis of Fab4F5 shows that the epitope interacts with the periphery of the hydrophobic pocket and provides insights into the molecular basis of mAb4F5 recognition and tight binding of FVIIaDVQ. CONCLUSION: The binary complex explains and supports the selectivity and functional consequences of Fab4F5 association with FVIIaDVQ and illustrates the potentially unique antigenicity of this FVIIa variant. This will be useful in the design of less immunogenic variants.

4.
Biophys J ; 116(10): 1823-1835, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31003762

ABSTRACT

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.


Subject(s)
Factor VIIa/chemistry , Factor VIIa/metabolism , Allosteric Regulation , Apoenzymes/chemistry , Apoenzymes/metabolism , Benzamidines/pharmacology , Crystallography, X-Ray , Disulfides/chemistry , Enzyme Activation/drug effects , Models, Molecular , Protein Domains , Protein Folding , Trypsin/chemistry , Trypsin/metabolism , Trypsinogen/metabolism
5.
Res Pract Thromb Haemost ; 3(1): 114-120, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30656284

ABSTRACT

BACKGROUND: Factor VIII (FVIII) procoagulant activity is commonly assessed by measuring the activated partial thromboplastin time (APTT) to clot formation using one of the many available but differently composed reagents. The majority of APTT reagents also accurately recover the activity of the extended half-life molecule N-glycoPEGylated FVIII (N8-GP; turoctocog alfa pegol), while a few silica-based reagents give a low recovery. OBJECTIVE: To identify the cause of N8-GP activity underestimation in the presence of certain silica-based APTT reagents. METHODS: Development of FVIIIa-dependent tenase activity and appearance of FVIIIa from N8-GP and its non-PEGylated counterpart (N8) were compared using clotting assays, a factor Xa (FXa) activity assay mimic thereof, and thrombin activation time courses. RESULTS: A strong correlation was demonstrated between clotting and FXa activity assays based on similar recoveries of N8-GP activity and equal responses to an altered duration of the contact activation phase, validating the latter as a useful clotting assay mimic. Contact activation phase duration influenced, and could even eliminate, N8-GP activity underestimation. Thrombin-catalyzed conversion of N8-GP to FVIIIa was considerably slower than that of N8 despite similar extents of adsorption to silica particles in APTT-SP, suggesting different modes and/or orientations of adsorption. CONCLUSIONS: Some contact activators reduce thrombin's ability to cleave N8-GP more than native FVIII. Decelerated thrombin activation of N8-GP is reflected in delayed FVIIIa-dependent appearance of FXa activity in plasma, in turn leading to prolonged clotting time. This forms the basis for underestimation of N8-GP activity as measured by one-stage clotting assay against a FVIII standard.

6.
Haemophilia ; 25(1): 154-161, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30664825

ABSTRACT

N9-GP (nonacog beta pegol; Refixia® ; Rebinyn® , Novo Nordisk A/S, Bagsvaerd, Denmark) is a glycoPEGylated extended half-life recombinant factor IX (rFIX) that exhibits efficacy and potency comparable to unmodified FIX molecules in non-clinical models. Phase 3 clinical trials have confirmed the efficacy and tolerability of N9-GP for the prevention and on-demand treatment of bleeding episodes in patients with haemophilia B. Recent studies have shown that PEGylation affects clotting times in activated partial thromboplastin time (aPTT)-based one-stage activity assays due to interaction between the FIX molecule and certain aPTT reagents. In recognition of the challenges surrounding FIX activity assessment, the identification of consistent, reproducible and accurate assays to measure FIX activity has been a priority for Novo Nordisk, running in parallel to the clinical development program for N9-GP. N9-GP activity can be reliably measured using chromogenic substrate assays and specific aPTT reagents. The conjugation of the PEG moiety to the FIX molecule may affect one-stage aPTT-based clotting assays in a reagent-dependent manner. Many aPTT reagents that use silica as the contact activator dramatically overestimate N9-GP activity due to premature activation. On the other hand, the contact activator in some other aPTT reagents negatively affects the enzymatic activity of FXIa, causing the underestimation of N9-GP activity. While N9-GP activity cannot be measured consistently with all available aPTT reagents, accurate N9-GP measurements can be achieved with certain aPTT reagents. Here, we review the studies that led to these findings and summarize the current options for accurate measurement of N9-GP in patient samples.


Subject(s)
Blood Coagulation Tests/methods , Factor IX/analysis , Polyethylene Glycols/analysis , Drug Monitoring , Factor IX/therapeutic use , Hemophilia B/drug therapy , Humans , Partial Thromboplastin Time , Polyethylene Glycols/therapeutic use , Recombinant Proteins/analysis , Recombinant Proteins/therapeutic use
8.
Res Pract Thromb Haemost ; 1(2): 259-263, 2017 Oct.
Article in English | MEDLINE | ID: mdl-30046695

ABSTRACT

BACKGROUND: In clinical practice, factor IX (FIX) activity is routinely quantified by measurement of the activated partial thromboplastin time (APTT) in a one-stage (OS) FIX clotting assay. APTT reagents provide a contact activator and phospholipid surfaces required for triggering and sustaining the plasma clotting process. The large diversity in reagent components is reflected in the variable recovery of nonacog beta pegol (N9-GP; N-glycoPEGylated recombinant FIX) activity when assayed against a FIX standard. This variation warrants mechanistic studies and is plausibly attributable to the nature and amount of contact activator. OBJECTIVE: To identify the cause of the N9-GP activity underestimation observed with a heterogeneous group of APTT reagents. METHODS: Experiments mimicking the clotting phase (omitting the contact activation phase) of the OS assay, complemented by measurements of activated factor XI (FXIa) activity, were performed to characterize and explain the influence of APTT reagents/contact activators on the conversion of N9-GP and regular FIX (N9) to activated FIX (FIXa). RESULTS: In the presence of an intact underestimating APTT reagent or the isolated contact activator, clotting phase activation of N9-GP proceeded at a reduced rate compared with that of N9. APTT reagent and contact activator negatively affected the activity of FXIa, conceivably as a consequence of FXIa adsorption. Thus, activation of FIX apparently poses a greater steric challenge after polyethylene glycol (PEG) conjugation. CONCLUSIONS: Some OS clotting assay contact activators reduce FXIa-mediated activation of N9-GP to a larger degree than that of N9, causing underestimation of N9-GP activity of potential clinical significance.

9.
Thromb Res ; 135(3): 544-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25596769

ABSTRACT

The number of patients on antithrombotic treatment due to atrial fibrillation and venous thromboembolism is increasing fast due to an aging population. A growing proportion will be treated with novel oral anticoagulants, the first in clinical use was the direct oral thrombin inhibitor dabigatran (Pradaxa®). A small percentage of the patients on dabigatran will experience serious bleeding or be in need of urgent surgery. The aim of this study was to test the effects of different hemostatic agents in potentially reversing the anticoagulant effects in vitro in blood or platelet-rich plasma (PRP) spiked with dabigatran. Whole blood or PRP was spiked with the active substance dabigatran, 200 µg/L. We measured clotting time being induced by 1.4 pmol/L tissue factor using the instrument ReoRox2™ and initial clot growth velocity from a tissue factor covered surface using the instrument Thrombodynamics Analyzer T-2™. Dabigatran prolonged clotting time 5-fold but reduced clot growth velocity only slightly. The reversing effects of prothrombin complex concentrates (PCC), activated PCC (APCC) and recombinant activated factor VII (rFVIIa) were then tested. APCC (1.8 U/mL) reduced the prolonged clotting time by 1/3, rFVIIa (2 µg/L) only slightly (n = 10-20). The reduction was not significant using Mann-Whitney test but significant using t-test with Bonferronis' correction for multiple comparisons, whereas PCC (0.56 U/mL) had no effect on clotting time. APCC doubled initial clot growth velocity, although even more in the absence of dabigatran. In conclusion, APCC and rFVIIa, but not PCC, seem to reverse, at least partially, some effects of dabigatran on coagulation parameters. Systematic evaluation of case reports, registries and, ultimately, randomized clinical trials are needed to elucidate potential benefit for patients.


Subject(s)
Antithrombins/pharmacology , Blood Coagulation Factors/pharmacology , Blood Coagulation/drug effects , Dabigatran/pharmacology , Factor VIIa/pharmacology , Hemostatics/pharmacology , Humans , Platelet-Rich Plasma/drug effects , Recombinant Proteins/pharmacology
10.
J Biol Chem ; 289(51): 35388-96, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25344622

ABSTRACT

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.


Subject(s)
Deuterium Exchange Measurement/methods , Factor VIIa/chemistry , Mass Spectrometry/methods , Protein Structure, Tertiary , Allosteric Site , Amino Acid Sequence , Binding Sites , Calcium/chemistry , Calcium/metabolism , Catalytic Domain , Crystallography, X-Ray , Electron Transport , Factor VIIa/metabolism , Glycosylation , Humans , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Secondary , Thromboplastin/chemistry , Thromboplastin/metabolism
11.
Protein Sci ; 23(12): 1717-27, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25234571

ABSTRACT

Formation of the factor VIIa (FVIIa)-tissue factor (TF) complex triggers the blood coagulation cascade. Using a structure-based rationale, we investigated how the length of the linker region between the two epidermal growth factor (EGF)-like domains in FVIIa influences TF binding and the allosteric activity enhancement, as well as the interplay between the γ-carboxyglutamic acid (Gla)-containing and protease domains. Removal of two residues from the native linker was compatible with normal cofactor binding and accompanying stimulation of the enzymatic activity, as was extension by two (Gly-Ser) residues. In sharp contrast, truncation by three or four residues abolished the TF-mediated stabilization of the active conformation of FVIIa and abrogated TF-induced activity enhancement. In addition, FVIIa variants with short linkers associated 80-fold slower with soluble TF (sTF) as compared with wild-type FVIIa, resulting in a corresponding increase in the equilibrium dissociation constant. Molecular modeling suggested that the shortest FVIIa variants would have to be forced into a tense and energetically unfavorable conformation in order to be able to interact productively with TF, explaining our experimental observations. We also found a correlation between linker length and the residual intrinsic enzymatic activity of Ca(2+)-free FVIIa; stepwise truncation resulting in gradually higher activity with des(83-86)-FVIIa reaching the level of Gla-domainless FVIIa. The linker appears to determine the average distance between the negatively charged Gla domain and a structural element in the protease domain, presumably of opposite charge, and proximity has a negative impact on apo-FVIIa activity.


Subject(s)
EGF Family of Proteins/chemistry , Factor VIIa/chemistry , Factor VIIa/metabolism , Thromboplastin/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Surface Plasmon Resonance
12.
Blood ; 124(20): 3172-4, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25232061

ABSTRACT

Successful competition of activated factor VII (FVIIa) with zymogen factor VII (FVII) for tissue factor (TF) and loading of the platelet surface with FVIIa are plausible driving forces behind the pharmacological effect of recombinant FVIIa (rFVIIa) in hemophilia patients. Thrombin generation measurements in platelet-rich hemophilia A plasma revealed competition for TF, which potentially could reduce the effective (r)FVIIa:TF complex concentration and thereby attenuate factor Xa production. However, (auto)activation of FVII apparently counteracted the negative effect of zymogen binding; a small impact was observed at endogenous concentrations of FVII and FVIIa but was virtually absent at pharmacological amounts of rFVIIa. Moreover, corrections of the propagation phase in hemophilia A required rFVIIa concentrations above the range where a physiological level of FVII was capable to downregulate thrombin generation. These data strongly suggest that rFVIIa acts independently of TF in hemophilia therapy and that FVII displacement by rFVIIa is a negligible mechanistic component.


Subject(s)
Coagulants/therapeutic use , Factor VIIa/therapeutic use , Hemophilia A/drug therapy , Thrombin/metabolism , Thromboplastin/metabolism , Factor VII/metabolism , Factor VIII/metabolism , Hemophilia A/blood , Humans , Recombinant Proteins/therapeutic use , Thrombin/analysis
13.
Blood ; 121(11): 2108-16, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23335368

ABSTRACT

Frequent infusions of intravenous factor VIII (FVIII) are required to prevent bleeding associated with hemophilia A. To reduce the treatment burden, recombinant FVIII with a longer half-life was developed without changing the protein structure. FVIII-polyethylene glycol (PEG) conjugates were prepared using an enzymatic process coupling PEG (ranging from 10 to 80 kDa) selectively to a unique O-linked glycan in the FVIII B-domain. Binding to von Willebrand factor (VWF) was maintained for all conjugates. Upon cleavage by thrombin, the B-domain and the associated PEG were released, generating activated FVIII (FVIIIa) with the same primary structure and specific activity as native FVIIIa. In both FVIII- and VWF-deficient mice, the half-life was found to increase with the size of PEG. In vivo potency and efficacy of FVIII conjugated with a 40-kDa PEG (N8-GP) and unmodified FVIII were not different. N8-GP had a longer duration of effect in FVIII-deficient mouse models, approximately a twofold prolonged half-life in mice, rabbits, and cynomolgus monkeys; however, the prolongation was less pronounced in rats. Binding capacity of N8-GP on human monocyte-derived dendritic cells was reduced compared with unmodified FVIII, resulting in several-fold reduced cellular uptake. In conclusion, N8-GP has the potential to offer efficacious prevention and treatment of bleeds in hemophilia A at reduced dosing frequency.


Subject(s)
Factor VIII/therapeutic use , Hemophilia A/drug therapy , Polyethylene Glycols/therapeutic use , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Disease Models, Animal , Factor VIII/administration & dosage , Factor VIII/metabolism , Female , Glycosylation , Hemophilia A/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/metabolism , Rabbits , Rats , Rats, Sprague-Dawley , Time Factors , Treatment Outcome
14.
Semin Thromb Hemost ; 38(3): 274-81, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22510860

ABSTRACT

Vatreptacog alfa is a genetically engineered variant of recombinant factor VIIa (rFVIIa) containing three amino acid changes. Aspartic acid, valine, and glutamine residues replace valine, glutamic acid, and methionine at positions 158, 296, and 298, respectively. These substitutions result in considerable enhancement of the intrinsic (tissue factor-independent) capability to activate factor X and the downstream hemostatic events are consequently augmented. The beneficial effects of vatreptacog alfa have been demonstrated in numerous in vitro systems attempting to mimic hemophilia and corroborated in in vivo models. Vatreptacog alfa has successfully passed through phase 1 and 2 clinical trials and the molecule is currently being explored in phase 3 clinical trial for the treatment of bleedings in hemophilia patients with inhibitors. This article describes the proposed mechanism behind the increased activity and action of vatreptacog alfa and reviews available data, which suggest that vatreptacog alfa could be a valuable addition to the existing portfolio of treatment options for hemophilia patients with inhibitors.


Subject(s)
Factor VIIa/pharmacology , Hemophilia A/drug therapy , Animals , CHO Cells , Cricetinae , Factor VIIa/genetics , Genetic Engineering , Humans , Molecular Structure , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
15.
Thromb Res ; 129 Suppl 2: S51-3, 2012 May.
Article in English | MEDLINE | ID: mdl-22405049

ABSTRACT

In the intact hemostatic system, the amount of factor Xa needed for efficient blood coagulation is supplied by the complex between factors VIIIa and IXa. Because hemophilia A patients lack factor VIII and hemophilia B patients lack factor IX, they share a bleeding phenotype that has its root in a dramatically decreased ability to generate factor Xa. These patients are currently treated by replacement therapy with factor VIII and IX, respectively, or, in case they have developed neutralizing inhibitory antibodies against the replacing factor, with a bypassing agent such as factor VIIa (NovoSeven®) or FEIBA®. This review briefly describes a number of novel promising approaches currently in the discovery or clinical development phase aiming at increased factor Xa generation in hemophilia.


Subject(s)
Factor Xa/biosynthesis , Hemophilia A/blood , Hemophilia A/drug therapy , Humans , Phenotype
16.
J Biol Chem ; 287(12): 8994-9001, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22275370

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/metabolism , Factor VIIa/chemistry , Factor VIIa/metabolism , Allosteric Regulation , Animals , Antibodies, Monoclonal/chemistry , Binding Sites , Factor VIIa/genetics , Humans , Kinetics , Mice , Protein Binding , Thromboplastin/genetics , Thromboplastin/metabolism
17.
Biochem Biophys Res Commun ; 413(4): 545-9, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21924243

ABSTRACT

The apparent length of FVIIa in solution was estimated by a FRET analysis. Two fluorescent probes, fluorescein (Fl-FPR) and a rhodamine derivative (TMR), were covalently attached to FVIIa. The binding site of Fl-FPR was in the protease domain whereas TMR was positioned in the Gla domain, thus allowing a length measure over virtually the whole extension of the protein. From the FRET measurements, the distances between the two probes were determined to be 61.4 for free FVIIa and 65.5Å for FVIIa bound to soluble tissue factor (sTF). These seemingly short distances, compared to those anticipated based on the complex crystal structure, require that the probes stretch towards each other. Thus, the apparent distance from the FRET analysis was shown to increase with 4Å upon formation of a complex with sTF in solution. However, considering how protein dynamics, based on recent molecular dynamics simulations of FVIIa and sTF:FVIIa (Y.Z. Ohkubo, J.H. Morrissey, E. Tajkhorshid, J. Thromb. Haemost. 8 (2010) 1044-1053), can influence the apparent fluorescence signal our calculations indicated that the global average conformation of active-site inhibited FVIIa is nearly unaltered upon ligation to sTF. It is known from amidolytic activity measurements that Ca(2+) binding leads to activation of FVIIa, but we have for the first time directly demonstrated conformational changes in the environment of the active site upon Ca(2+) binding. Interestingly, this Ca(2+)-induced conformational change can be noted even in the presence of an inhibitor. Forming a complex with sTF further stabilized this conformational change, leading to a more inaccessible active-site located probe.


Subject(s)
Calcium/chemistry , Factor VIIa/chemistry , Thromboplastin/chemistry , Amino Acid Substitution , Catalytic Domain , Enzyme Activation , Factor VIIa/genetics , Fluorescence Resonance Energy Transfer , Protein Binding , Protein Conformation , Protein Structure, Tertiary
18.
Front Biosci (Landmark Ed) ; 16(8): 3156-63, 2011 06 01.
Article in English | MEDLINE | ID: mdl-21622226

ABSTRACT

Coagulation factor VIIa (FVIIa) is present at subnanomolar concentration and represents a small percentage of the total amount of FVII in the circulation. FVIIa is poised to initiate blood clotting when it encounters its pivotal cofactor tissue factor (TF) which becomes exposed to blood upon vascular rupture. The requirement for complex formation with TF in order for FVIIa to express procoagulant activity ensures thrombin and fibrin generation at the right time and place. Thus TF acts as a guardian of safety of paramount importance to blood coagulation by providing localization to the site of injury and at the same time inducing maturation of zymogen-like free FVIIa to the active cofactor-bound enzyme. This review gives an account of the accumulated knowledge about the structure, function and TF dependence of FVIIa to arrive at a plausible allosteric mechanism by which TF induces maturation of the active conformation of FVIIa.


Subject(s)
Factor VIIa/chemistry , Factor VIIa/metabolism , Allosteric Regulation , Animals , Blood Coagulation/physiology , Humans , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Conformation , Protein Structure, Tertiary , Thromboplastin/chemistry , Thromboplastin/metabolism
19.
Blood ; 117(12): 3445-52, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21252090

ABSTRACT

Coagulation factors VII (FVII), IX (FIX), X (FX), and protein C share the same domain organization but display very different plasma half-lives. It is plausible that the half-life is influenced by the activation peptide, differing in length and glycosylation and missing in FVII. To test this hypothesis, the influence of activation peptides on the plasma half-life of human FVII was studied by administering human FVII variants containing activation peptide motifs to mice. Insertion of the activation peptide from FX gave 4-fold longer terminal half-life (5.5 hours vs 1.4 hours for FVII), whereas the activation peptide from FIX and protein C resulted in half-lives of 4.3 and 1.7 hours, respectively. Using FX's activation peptide we identified the N-linked glycans as structural features important for the half-life. The peptide location within the FVII molecule appeared not to be critical because similar prolongation was obtained with the activation peptide inserted immediately before the normal site of activation and at the C-terminus. However, only the latter variant was activatable, yielding full amidolytic activity and reduced proteolytic activity with preserved long half-life. Our data support that activation peptides function as plasma retention signals and constitute a new manner to extend the half-life of FVII(a).


Subject(s)
Factor VII/chemistry , Factor VII/pharmacokinetics , Peptide Fragments/physiology , Plasma/metabolism , Amino Acid Motifs , Animals , Factor IX/chemistry , Factor IX/immunology , Factor IX/metabolism , Factor IX/pharmacokinetics , Factor VII/metabolism , Factor X/chemistry , Factor X/immunology , Factor X/metabolism , Factor X/pharmacokinetics , Half-Life , Humans , Male , Mice , Mutant Proteins/chemistry , Mutant Proteins/pharmacokinetics , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Peptide Fragments/immunology , Peptide Fragments/pharmacokinetics , Protein Processing, Post-Translational
20.
Br J Haematol ; 152(1): 99-107, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21083658

ABSTRACT

NN1731 is a recombinant activated factor VII (rFVIIa) analogue with increased intrinsic activity. This also applies to its reactivity towards antithrombin (AT), the role of which was investigated in a pharmacokinetic (PK) study. NN1731 or rFVIIa was administered to normal and haemophilia A dogs and elimination was measured by FVIIa clot activity, FVIIa- and FVIIa-AT antigen. In vitro AT complex formation was studied in canine plasma spiked with NN1731 or rFVIIa. Based on FVIIa antigen concentrations, PK profiles in normal and haemophilia A dogs were similar for NN1731 and rFVIIa with antigen half lives, t(½) ≈1·8 h. In contrast, PK profiles based on activity measurements were distinctly different. NN1731 induced a strong, short lasting (t(½) ≈0·5 h) pro-coagulant response, whereas rFVIIa induced a lower, longer lasting (t(½) ≈1·1 h) response. Western Blot and FVIIa-AT antigen analysis demonstrated in vivo AT complex formation that accounted for these divergences. AT complex formation with FVIIa or NN1731 in vitro in canine plasma was considerably slower than the in vivo reaction. The results suggest that in vivo inhibition by AT contributes significantly to define drug duration in haemophilia treatment with rFVIIa and in particular with the NN1731 analogue.


Subject(s)
Antithrombin Proteins/physiology , Coagulants/pharmacokinetics , Factor VII/pharmacokinetics , Hemophilia A/blood , Animals , Blood Coagulation/drug effects , Blood Coagulation Factor Inhibitors/physiology , Coagulants/antagonists & inhibitors , Disease Models, Animal , Dogs , Factor VII/antagonists & inhibitors , Factor VIIa/antagonists & inhibitors , Half-Life , Male , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/blood , Thrombelastography/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...