scholarly journals BFPI: A Minimal Prothrombinase Inhibitor from the Saliva of Simulium Vittatum

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1163-1163
Author(s):  
Jeremy P. Wood ◽  
Xiao H. Song ◽  
Alan E. Mast

Abstract Background: The black fly, Simulium Vittatum, has an anticoagulant protein in its saliva that allows it to feed on mammalian blood (black fly protease inhibitor; BFPI). Remarkably, BFPI is similar to the human anticoagulant tissue factor pathway inhibitor alpha (TFPIα). TFPIα contains three Kunitz-type protease inhibitor domains (K1, K2, K3), which inhibit factor VIIa (FVIIa) and factor Xa (FXa) and bind the co-factor protein S (PS), respectively; BFPI contains a single Kunitz domain that inhibits FXa. In addition, TFPIα and BFPI contain homologous basic regions (BRs) near their C-termini (252LIKTKRKRKK261 in human TFPIα, LIKTRKRKPKK in BFPI). The TFPIα BR binds a regulatory acidic region (AR) in factor Va (FVa). The AR is present in forms of FVa released by collagen-activated platelets and generated through limited proteolysis by FXa (FVaXa), and is removed by thrombin (FVaIIa). We hypothesized that BFPI, through its Kunitz domain and basic C-terminus, inhibits early forms of the prothrombinase complex, but does not possess the other inhibitory functions of TFPIα: (1) K1-dependent inhibition of the tissue factor (TF)-FVIIa complex; and (2) PS/K3-dependent FXa inhibition. Results: Recombinant BFPI inhibited FXa in an amidolytic activity assay, and PS did not promote this inhibition. BFPI did not inhibit TF-FVIIa-mediated FX activation. As described with TFPIα, FV promoted FXa inhibition by BFPI but FVaIIa did not, suggesting that the BFPI BR is capable of binding the FVa AR. In a purified protein assay, BFPI inhibited prothrombinase assembled with FVaXa (IC50=4.9nM), but not FVaIIa. Similarly, 5nM BFPI increased the lag time for FXa-initiated plasma thrombin generation by 10.4±1.5%. We next used BFPI as a backbone to evaluate a reported human mutation in the TFPIα BR, K254E. Every mammalian, avian, or reptilian TFPIα sequence available contains either a Lys or Arg residue at this position, suggesting that this residue is functionally important. In purified protein assays, BFPI-K254E inhibited FXa amidolytic activity identically to BFPI, but FV did not promote this inhibition, suggesting that BFPI-K254E has a specific defect in its interaction with FV. Consistent with this, BFPI-K254E was a weaker inhibitor of prothrombinase assembled with FVaXa (IC50 = 15.8nM) and FXa-initiated plasma thrombin generation. The results obtained with BFPI-K254E were confirmed using peptides and full-length TFPIα proteins. First, a peptide mimicking the wild type TFPIα BR (LIKTKRKRKK) inhibited prothrombinase assembled with FVaXa (IC50 = 1.0 µM), while the substituted peptide (LIETKRKRKK) was substantially weaker (20% inhibition observed with 340 µM peptide). Second, full-length TFPIα-K254E was a weaker inhibitor of prothrombinase containing FXa-activated FVa (IC50 = 14.8 nM, vs. 1.8 nM for TFPIα) and had greatly reduced anticoagulant activity in plasma-based thrombin generation assays. Conclusions: In summary, the anticoagulant effect of BFPI is mediated through inhibition of early forms of prothrombinase, independent of TF-FVIIa inhibition or PS-dependent FXa inhibition. The natural mutation TFPIα K254E disrupts prothrombinase inhibition, despite the presence of six other conserved basic residues, and is thus procoagulant in human plasma. The absolute conservation of the TFPIα BR, and its usurpation to allow feeding by black flies, point to formation of the initial prothrombinase complex as a key regulatory step in blood coagulation. Disclosures Mast: Novo Nordisk: Research Funding.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4417-4417 ◽  
Author(s):  
Michael Palige ◽  
Christoph Redl ◽  
Sabine Knappe ◽  
Hartmut J. Ehrlich ◽  
Michael Dockal ◽  
...  

Abstract Abstract 4417 BAX513, a fucoidan derived from the brown seaweed Laminaria japonica, and other non-anticoagulant sulfated polysaccharides (NASPs) improve coagulation in hemophilic blood and plasma. Fucoidans are heterogeneous, polysulfated molecules with procoagulant activities in a wide concentration range. Tissue factor pathway inhibitor (TFPI) has been described as a potential target for the procoagulant activity of NASPs (Liu et al. Thromb Haemost 2006; 95:68). In the current study, we investigated the interaction of BAX513 with TFPI proteins to gain a detailed understanding of the mechanism of action of BAX513. We used calibrated automated thrombography to monitor the activity of BAX513 in normal, FX and TFPI-deficient plasma. TFPI plasma levels were varied by the addition of truncated TFPI (TFPI1-160) and TFPI-domain specific antibodies. Initiating thrombin generation by addition of FXa to plasma deficient in both, FX and FVIII-showed a BAX513-dose dependent increase of thrombin generation, which was completely abolished when TFPI-specific polyclonal antibodies were present. Furthermore, when full-length TFPI was inhibited in plasma and instead supplemented with increasing amounts of TFPI 1–160, BAX513 did not show any activity. The data are further supported by surface plasmon resonance experiments (BiaCore) exploring the BAX513-TFPI interaction. A high affinity interaction was only observed for BAX513 with full-length TFPI but not for BAX513 with TFPI1-160. Our findings support a mechanism of action in which BAX513 acts as a potent dose-dependent TFPI antagonist that requires the highly charged C-terminus of TFPI to unfold its full potential. Understanding the mechanism of action of BAX513 supports the development of BAX513 as a promising new therapeutic for hemophiliacs and FVIII-inhibitor patients. Disclosures: Palige: Baxter Innovations GmbH: Employment. Redl:Baxter Innovations GmbH: Employment. Knappe:Baxter Innovations GmbH: Employment. Ehrlich:Baxter Innovations GmbH: Employment. Dockal:Baxter Innovations GmbH: Employment. Scheiflinger:Baxter Innovations GmbH: Employment.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2245-2245
Author(s):  
Michael Dockal ◽  
Johannes Brandstetter ◽  
Martin Ludwiczek ◽  
Georg Kontaxis ◽  
Markus Fries ◽  
...  

Abstract Abstract 2245 Blood coagulation is initiated by the tissue factor-factor VIIa (TF-FVIIa) complex which cleaves and activates coagulation factor X to Xa (FXa). Tissue factor pathway inhibitor (TFPI) controls this key process and thus plays a crucial role in maintaining the delicate balance of pro- and anticoagulant processes. Inhibition of TFPI in hemophilia plasma and in a rabbit model of hemophilia has been shown to improve coagulation and hemostasis (Nordfang et al., Thromb Haemost. 1991;66:464; Erhardsen et al., Blood Coagulation and Fibrinolysis 1995;6:388). TFPI is a Kunitz-type protease inhibitor that inhibits FXa and TF-FVIIa. TFPI is a slow, tight-binding FXa inhibitor which rapidly forms a loose FXa-TFPI complex that slowly isomerises to a tight FXa-TFPI* complex. The FXa-TFPI* complex inhibits TF-FVIIa by formation of a quaternary FXa-TFPI-TF-FVIIa complex. Using a library approach, we selected a peptide which binds and inhibits TFPI. We located the binding site of the antagonistic peptide on TFPI by NMR spectroscopy. Residues of TFPI undergoing the strongest chemical shift changes were exclusively found on the Kunitz domain 1 (KD1). NMR data were confirmed by solving the crystal structure of KD1 in complex with the antagonistic peptide at 2.55 Å resolution. Like in related Kunitz domains, the robustness of this approximately 60-amino-acid long folding module largely depends on stabilization by the three disulfides bonds and a hydrophobic cluster of three phenylalanines. The disulfide bridging of the P2 residue induces conformational constraints on the reactive centre loop (RCL), thereby establishing an extended RCL conformation; consequently, the amino acid side chains flanking the “scissile” peptide bond are exposed to the solvent. This RCL geometry also explains why the distorted, improperly activated scissile peptide bond is hardly cleaved. Whereas Cys-Lys/Arg is a rather conserved P2-P1 motif, reflecting the topological restraints in Kunitz protease inhibitors, proline at position P3 induces an additional conformational constraint on the RCL, which would not be possible in the narrow active site of FXa. Proline at the P3 and to a lesser extent Lys rather than Arg at P1 thus represent two major specificity determinants of KD1 towards FVIIa over FXa. The structure of the 20-mer peptide can be segmented into (i) an N-terminal anchor; (ii) an Ω-shaped loop; (iii) an intermediate segment; (iv) a tight glycine loop; and (v) a C-terminal α-helix that is anchored to KD1 at its RCL and two-strand β-sheet. The contact surface has an overall hydrophobic character with some charged hot spots but the major driving force of complex formation is steric surface complementarity. One of the optimized peptides, which binds to KD1 of TFPI, had an affinity for TFPI of <1 nM. In a model system, the peptide blocked both FXa inhibition by TFPI (IC50=5 nM) and inhibition of TF-FVIIa-catalyzed FX activation by TFPI (IC50=5.7 nM). In FVIII-depleted plasma, the peptide enhanced thrombin generation 9-fold (EC50=4 nM). Detailed kinetic analysis in a model system showed that the peptide almost fully inhibited TFPI and prevented the transition from the loose to the tight FXa-TFPI* complex, but did not affect formation of the loose FXa-TFPI complex. Since KD1 binds to the active site of FVIIa and KD2 to the active site of FXa our kinetic data with the KD1-binding peptide show that KD1 is not only important for FVIIa inhibition but is also required for FXa inhibition, i.e. for the transition from the loose to the tight FXa-TFPI* complex. In line with this mechanism, the peptide did not affect FXa inhibition by the isolated KD2. The peptide was also able to dissociate preformed FXa-TFPI* and FXa-TFPI-TF-FVIIa complexes and liberate active FXa and TF-FVIIa. In summary, we developed a peptide that binds to KD1 of TFPI, that prevents FXa-TFPI and FXa-TFPI-TF-FVIIa complex formation and that enhances coagulation under hemophilia conditions. Disclosures: Dockal: Baxter Innovations GmbH: Employment. Brandstetter:University of Salzburg: Employment. Ludwiczek:Baxter Innovations GmbH: Employment. Kontaxis:University of Vienna: Employment. Fries:Baxter Innovations GmbH: Employment. Thomassen:Maastricht University: Employment. Heinzmann:Maastricht University: Employment. Ehrlich:Baxter Innovations GmbH: Employment. Prohaska:Baxter Innovations GmbH: Employment. Hartmann:Baxter Innovations GmbH: Employment. Rosing:Maastricht University: Employment. Scheiflinger:Baxter Innovations GmbH: Employment.


2013 ◽  
Vol 394 (7) ◽  
pp. 819-829 ◽  
Author(s):  
Saulius Butenas

AbstractTissue factor (TF), an initiator of blood coagulationin vivo, is expressed in a variety of cells. Sufficient natural TF has been isolated to clone and express recombinant proteins ranging from full-length TF to its extracellular domain. Because of the limited availability of natural TF, recombinant proteins have been used as surrogates. Despite the differences in their post-translational modifications, it has been accepted that membrane-anchored recombinant TFs are quite similar to the natural TF. Recent studies, however, have shown that post-translational modifications play an important role in TF-triggered thrombin generation.


Blood ◽  
2011 ◽  
Vol 118 (2) ◽  
pp. 437-445 ◽  
Author(s):  
Anton Matafonov ◽  
Suryakala Sarilla ◽  
Mao-fu Sun ◽  
John P. Sheehan ◽  
Vladimir Serebrov ◽  
...  

AbstractThe prothrombinase complex converts prothrombin to α-thrombin through the intermediate meizothrombin (Mz-IIa). Both α-thrombin and Mz-IIa catalyze factor (F) XI activation to FXIa, which sustains α-thrombin production through activation of FIX. The interaction with FXI is thought to involve thrombin anion binding exosite (ABE) I. α-Thrombin can undergo additional proteolysis to β-thrombin and γ-thrombin, neither of which have an intact ABE I. In a purified protein system, FXI is activated by β-thrombin or γ-thrombin, and by α-thrombin in the presence of the ABE I-blocking peptide hirugen, indicating that a fully formed ABE I is not absolutely required for FXI activation. In a FXI-dependent plasma thrombin generation assay, β-thrombin, γ-thrombin, and α-thrombins with mutations in ABE I are approximately 2-fold more potent initiators of thrombin generation than α-thrombin or Mz-IIa, possibly because fibrinogen, which binds to ABE I, competes poorly with FXI for forms of thrombin lacking ABE I. In addition, FXIa can activate factor FXII, which could contribute to thrombin generation through FXIIa-mediated FXI activation. The data indicate that forms of thrombin other than α-thrombin contribute directly to feedback activation of FXI in plasma and suggest that FXIa may provide a link between tissue factor-initiated coagulation and the proteases of the contact system.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 497-497 ◽  
Author(s):  
Sabine Knappe ◽  
Nicholas Kripalani ◽  
Susannah Patarroyo-White ◽  
Kirk W. Johnson ◽  
Srinivasa Prasad

Abstract AV513, a sulfated polysaccharide prepared from brown seaweed and enriched for fucoidan accelerates clotting in hemophilia A and B plasma. Prior studies have demonstrated that AV513 functions mainly by inhibiting TFPI activity. In this report, we have further studied AV513 mechanism of action using human plasma thromboelastography (TEG) model and a thrombin generation assay in FVIII deficient plasma. In the TEG assay, AV513 enhanced tissue factor (TF) induced clotting in human hemophilia A pooled plasma in a dose-dependent manner. A dose-dependent potentiation effect by AV513 was observed with reduction of TEG R time to normal levels at 0.5 μg/mL of AV513. An AV513 dose dependent increase in thrombin generation was observed in hemophilia A pooled plasma with a 3-fold increase in peak thrombin generation at 2.5 μg/mL AV513 (basal: 40 nM vs. treated: 140 nM). When TFPI was immunodepleted from hemophilia A plasma (FVIII/TFPI deficient), sub-optimal concentrations of TF enhanced thrombin generation which was not further enhanced by addition of AV513. The increase in thrombin generation in FVIII /TFPI deficient plasma was inhibited by 93% after the addition of exogenous full-length TFPI or by 47% after the addition of truncated rTFPI (amino acids 1–161) that lacks the Kunitz 3 domain and the C-terminal tail. AV513 at low concentrations reversed exogenous full length TFPI activity almost completely in FVIII/TFPI-deficient plasma but had only a small effect on truncated TFPI. In a human plasma TEG combination study of AV513 and low concentrations of rFVIII (ReFacto), AV513 at a concentration of 0.25 μg/mL reduced the TEG R time by 35% in severe hemophilia A plasma compared to 10 mU/mL of rFVIII alone. These results indicate that AV513 functions as a pro-coagulant by inhibiting TFPI at least in part via targeting the C-terminal region. Moreover, AV513, in the presence of low rFVIII (ReFacto) levels, improves severe hemophilia A plasma clotting - as measured by TEG - beyond that observed with either agent alone.


1997 ◽  
Vol 323 (1) ◽  
pp. 33-37 ◽  
Author(s):  
Jo FRANSSEN ◽  
Irene SALEMINK ◽  
George M. WILLEMS ◽  
Tze-Chein WUN ◽  
H. Coenraad HEMKER ◽  
...  

The inhibition of prothrombinase by tissue factor pathway inhibitor (TFPI) has been studied in the presence and absence of prothrombin. The rate constant of association of prothrombinase with full-length TFPI was 2.1×107 M-1ċs-1 and 0.05×107 M-1ċs-1 for the reaction with C-terminus truncated TFPI (TFPI1-161). The rate constant of dissociation was 0.65×10-4 s-1 in both cases. The rate constant of inhibition of prothrombinase by TFPI1-161 was similar to that of solution-phase factor Xa. In contrast, phospholipids and factor Va enhanced the association rate of the reaction between factor Xa and full-length TFPI by approx. 20-fold. Although TFPI, and in particular the full-length variant of the molecule, is a potent inhibitor of prothrombinase (overall inhibition constant of 3 pM), we also found that prothrombin competed very effectively with TFPI for the active site of factor Xa in the prothrombinase complex. A 50% reduction of the rate constant of inhibition was measured in the presence of 4 nM prothrombin, i.e. 0.2% of the plasma concentration of prothrombin. The physiological significance of TFPI as an inhibitor of prothrombinase activity is thus questionable.


TH Open ◽  
2021 ◽  
Vol 05 (03) ◽  
pp. e363-e375
Author(s):  
Ton Lisman ◽  
Jelle Adelmeijer ◽  
Dana Huskens ◽  
Joost C. M. Meijers

Abstract Background Aprotinin is a broad-acting serine protease inhibitor that has been clinically used to prevent blood loss during major surgical procedures including cardiac surgery and liver transplantation. The prohemostatic properties of aprotinin likely are related to its antifibrinolytic effects, but other mechanisms including preservation of platelet function have been proposed. Aim Here we assessed effects of aprotinin on various hemostatic pathways in vitro, and compared effects to tranexamic acid(TXA), which is an antifibrinolytic but not a serine protease inhibitor. Methods We used plasma-based clot lysis assays, clotting assays in whole blood, plasma, and using purified proteins, and platelet activation assays to which aprotinin or TXA were added in pharmacological concentrations. Results Aprotinin and TXA dose-dependently inhibited fibrinolysis in plasma. Aprotinin inhibited clot formation and thrombin generation initiated via the intrinsic pathway, but had no effect on reactions initiated by tissue factor. However, in the presence of thrombomodulin, aprotinin enhanced thrombin generation in reactions started by tissue factor. TXA had no effect on coagulation. Aprotinin did not inhibit thrombin, only weakly inhibited the TF-VIIa complex and had no effect on platelet activation and aggregation by various agonists including thrombin. Aprotinin and TXA inhibited plasmin-induced platelet activation. Conclusion Pharmacologically relevant concentrations of aprotinin inhibit coagulation initiated via the intrinsic pathway. The antifibrinolytic activity of aprotinin likely explains the prohemostatic effects of aprotinin during surgical procedures. The anticoagulant properties may be beneficial during surgical procedures in which pathological activation of the intrinsic pathway, for example by extracorporeal circuits, occurs.


1993 ◽  
Vol 70 (03) ◽  
pp. 454-457 ◽  
Author(s):  
Claus Bregengaard ◽  
Ole Nordfang ◽  
Per Østergaard ◽  
Jens G L Petersen ◽  
Giorgio Meyn ◽  
...  

SummaryTissue factor pathway inhibitor (TFPI) is a feed back inhibitor of the initial activation of the extrinsic pathway of coagulation. In humans, injection of heparin results in a 2-6 fold increase in plasma TFPI and recent studies suggest that TFPI may be important for the anticoagulant activity of heparin. Full length (FL) TFPI, but not recombinant two-domain (2D) TFPI, has a poly cationic C-terminus showing very strong heparin binding. Therefore, we have investigated if heparin affects the pharmacokinetics of TFPI with and without this C-terminus.FL-TFPI (608 U/kg) and 2D-TFPI (337 U/kg) were injected intravenously in rabbits with and without simultaneous intravenous injections of low molecular weight heparin (450 anti-XaU/kg).Heparin decreased the volume of distribution and the clearance of FL-TFPI by a factor 10-15, whereas the pharmacokinetics of 2D-TFPI were unaffected by heparin. When heparin was administered 2 h following TFPI the recovery of FL-TFPI was similar to that found in the group receiving the two compounds simultaneously, suggesting that the releasable pool of FL-TFPI is removed very slowly in the absence of circulating heparin.


1993 ◽  
Vol 69 (03) ◽  
pp. 227-230 ◽  
Author(s):  
J Van Ryn-McKenna ◽  
H Merk ◽  
T H Müller ◽  
M R Buchanan ◽  
W G Eisert

SummaryWe compared the relative abilities of unfractionated heparin and annexin V to prevent fibrin accretion onto injured jugular veins in vivo. Heparin was used to accelerate the inhibition of thrombin by antithrombin III, and annexin V was used to inhibit the assembly of the prothrombinase complex on phospholipid surfaces, thereby blocking thrombin generation. Rabbit jugular veins were isolated in situ, a 2 cm segment was injured by perfusing it with air, and then blood flow was re-established. Five minutes later, each rabbit was injected with heparin (20 U/kg) or annexin V (0.3 mg/kg) and then with 125I-fibrinogen. The amount of 125I-fibrin accumulation onto each injured vessel wall segment was measured 4 h later. Each injured vessel was completely deendothelialized as a result of the air perfusion as demonstrated by electron microscopy. 125I-fibrin accretion onto the injured jugular veins was enhanced 2.4-fold as compared to the uninjured veins in sham-operated animals. Heparin treatment did not reduce fibrin accretion, whereas, annexin V treatment decreased fibrin accretion by 60%, p <0.05. This latter effect was achieved without sustained circulating anticoagulation. Additional experiments confirmed that the inhibitory effect of annexin V on fibrin accretion was associated with a surface specific effect, since more annexin V bound to the injured jugular vein segments as compared to the non-injured jugular veins. We conclude that, i) mild vessel wall injury (selective de-endothelialization) in veins results in a thrombogenic vessel wall; ii) the thrombogenecity of which is not inhibited by prophylactic doses of heparin; but iii) is inhibited by annexin V, which binds to injured vessel wall surface, and inhibits thrombin generation independently of antithrombin III.


Sign in / Sign up

Export Citation Format

Share Document