scholarly journals Plasma protein S contains zinc essential for efficient activated protein C‐independent anticoagulant activity and binding to factor Xa, but not for efficient binding to tissue factor pathway inhibitor

2009 ◽  
Vol 23 (7) ◽  
pp. 2244-2253 ◽  
Author(s):  
Mary J. Heeb ◽  
Duane Prashun ◽  
John H. Griffin ◽  
Bonno N. Bouma
2011 ◽  
Vol 106 (11) ◽  
pp. 914-921 ◽  
Author(s):  
M. Christella Thomassen ◽  
Serban-Dan Costa ◽  
Louis Peeters ◽  
Jan Rosing ◽  
Svetlana Tchaikovski

SummaryPregnancy increases the risk of venous thromboembolism. Particularly in early pregnancy, the thrombosis risk can be attributed to the changes in coagulation. Elevated thrombin generation and resistance to activated protein C (APC) are likely to contribute to the increased thrombosis risk during pregnancy. We studied changes and the determinants of thrombin generation and APC resistance in the first 16 weeks of gestation in women with history of preeclampsia. Additionally, we investigated the influence of pregnancy-induced haemodilution on the coagulation system. We measured thrombin generation, APC resistance and plasma levels of prothrombin, factor V, factor X, protein S and tissue factor pathway inhibitor (TFPI) in 30 non-pregnant and 21 pregnant women at 8, 12 and 16 weeks of gestation. All participants shared a history of a hypertensive complication in the preceding pregnancy. Thrombin generation and APC resistance were higher at eight weeks of pregnancy than in the non-pregnant state, and progressively increased between eight and 16 weeks of gestation. Changes in the TFPI and protein S levels accounted for ~70% of pregnancy-induced APC resistance. Interestingly, a significant correlation (slope 2.23; 95%CI: 1.56 to 2.91; r= 0.58) was observed between protein Stotal or protein Sfree levels and haematocrit. In conclusion, pregnancy induces a decrease of TFPIfree and protein Sfree levels that attenuates the function of the TFPI and protein C systems and results in elevated thrombin generation and increased APC resistance. Besides, our data suggest that pregnancy-dependent haemodilution may contribute to the decreased peripheral protein S levels.


2016 ◽  
Vol 36 (suppl_1) ◽  
Author(s):  
Jeremy P Wood ◽  
Lisa M Baumann Kreuziger ◽  
Susan A Maroney ◽  
Rodney M Camire ◽  
Alan E Mast

Factor V (FV) assembles with factor Xa (FXa) into prothrombinase, the enzymatic complex that converts prothrombin to thrombin. Tissue factor pathway inhibitor α (TFPIα) inhibits prothrombinase by high affinity interactions with FXa-activated FV and the FXa active site, thereby blocking the initiation of coagulation. FV Leiden (FVL) is strongly linked to venous thrombosis through its resistance to degradation by activated protein C (aPC), which enhances the propagation of coagulation. FVL combined with a 50% reduction in TFPI causes severe thrombosis and perinatal lethality in mice, suggesting that FVL also promotes the initiation of coagulation. To examine this possibility, thrombin generation assays initiated with limiting FXa were performed with control or FVL plasma and platelet-rich plasma (PRP). The activation threshold for thrombin generation was 10 to 20 pM FXa in 10 control plasmas, but was 5 pM in 4 of 10 homozygous FVL plasmas. FVL PRP had a similar decrease in the activation threshold. The differences in activation threshold were totally normalized by an anti-TFPI antibody, while exogenous TFPIα and a FV-binding peptide that mimics TFPIα had reduced anticoagulant activity in FVL plasma, revealing that the procoagulant effects of FVL in these assays rely on TFPIα. Next, FVL plasmas were studied in fibrin clot formation assays, as they are sensitive to small amounts of thrombin. In reactions activated with 0.5 pM FXa, 1 of 8 control plasmas, compared to 7 of 8 homozygous FVL plasmas, clotted within 60 minutes, with differences again normalized by the anti-TFPI antibody. In prothrombinase activity assays using purified proteins, TFPIα was a 1.7-fold weaker inhibitor of prothrombinase assembled with FVL compared to FV. Thus, in addition to its aPC-mediated effect on the propagation of coagulation, FVL is resistant to TFPIα inhibition, exerting a procoagulant effect on coagulation initiation. This is evident in responses to small stimuli, where TFPIα blocks clotting in plasmas with FV but not FVL. The TFPIα-mediated modulation of the procoagulant threshold may explain the severe perinatal thrombosis in FVL mice with decreased TFPI and be clinically relevant in the clotting associated with oral contraceptives, which cause acquired TFPI deficiency.


2001 ◽  
Vol 360 (2) ◽  
pp. 499-506 ◽  
Author(s):  
Delphine BORGEL ◽  
Pascale GAUSSEM ◽  
Christiane GARBAY ◽  
Christilla BACHELOT-LOZA ◽  
Tahar KAABACHE ◽  
...  

In the vitamin K-dependent protein family, only protein S (PS) contains a thrombin-sensitive region (TSR), located between the domain containing the γ-carboxyglutamic acid and the first epidermal growth factor-like domain. To better define the role of TSR in the PS molecule, we expressed a recombinant human PS (rHPS) and its analogue lacking TSR (rTSR-less), and prepared factor Xa- and thrombin-cleaved rHPS. A peptide reproducing TSR (TSR-peptide) was also synthesized in an attempt to obtain direct evidence of the domain involvement in PS anticoagulant activity. In a coagulation assay, both rTSR-less and factor Xa-cleaved PS were devoid of activated protein C cofactor activity. The TSR-peptide did not inhibit rHPS activity, showing that TSR must be embedded in the native protein to promote interaction with activated protein C. The binding of rHPS to activated platelets and to phospholipid vesicles was not modified after factor Xa- or thrombin-mediated TSR cleavage, whereas the binding of rTSR-less was markedly reduced. This suggested a role for TSR in conferring to PS a strong affinity for phospholipid membranes. TSR-peptide did not directly bind to activated platelets or compete with rHPS for phospholipid binding. The results of the present study show that TSR may not interact directly with membranes, but probably constrains the γ-carboxyglutamic acid-rich domain in a conformation allowing optimal interaction with phospholipids.


2013 ◽  
Vol 289 (3) ◽  
pp. 1732-1741 ◽  
Author(s):  
Michael Dockal ◽  
Rudolf Hartmann ◽  
Markus Fries ◽  
M. Christella L. G. D. Thomassen ◽  
Alexandra Heinzmann ◽  
...  

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 β-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.


Author(s):  
Adrienn Teraz-Orosz ◽  
Magdalena Gierula ◽  
Anastasis Petri ◽  
David A. Jones ◽  
Renos Keniyopoullos ◽  
...  

Protein S is a cofactor in the tissue factor pathway inhibitor (TFPI) anticoagulant pathway. It enhances TFPIα-mediated inhibition of factor (F)Xa activity and generation. The enhancement is dependent on a TFPIα-protein S interaction, involving TFPIα Kunitz 3 and protein S laminin G-type (LG)-1. C4b binding protein (C4BP), which binds to protein S LG1, almost completely abolishes its TFPI cofactor function. However, neither the amino acids involved in TFPIα enhancement, nor the mechanisms underlying the reduced TFPI cofactor function of C4BP-bound protein S, are known. To screen for functionally important regions within protein S LG1 we generated seven variants with inserted N-linked glycosylation attachment sites. Protein S D253T and Q427N/K429T, displayed severely reduced TFPI cofactor function while showing normal activated protein C (APC) cofactor function and C4BP binding. Based on these results, we designed four protein S variants in which 4-6 surface exposed charged residues were substituted for alanine. One variant, protein S K255A/E257A/D287A/R410A/K423A/E424A, exhibited either abolished or severely reduced TFPI cofactor function in plasma and FXa inhibition assays, both in the presence or absence of FV-short, but retained normal APC cofactor function and high affinity C4BP-binding. The C4BP β-chain was expressed to determine the mechanisms behind the reduced TFPI cofactor function of C4BP-bound protein S. Like C4BP-bound protein S, C4BP β-chain-bound protein S had severely reduced TFPI cofactor function. These results show that protein S Lys255, Glu257, Asp287, Arg410, Lys423 and Glu424 are critical for protein S-mediated enhancement of TFPIα and that binding of the C4BP β-chain blocks this function.


1981 ◽  
Author(s):  
F J Walker

The anticoagulant activity of activated Protein C has been observed to be species specific. This could be due either to the inability of the bovine enzyme to recognize its substrate, Factor Va, in non-bovine plasmas, or the absence of cofactor-Protein S, a protein that has been shown to be necessary for the maximum expression of the anticoagulant activity of activated Protein C. Activated Protein C was found to be an effective inhibitor of Factor Xa-initiated clotting of bovine plasma, but without activity in either human or rabbit plasma. Human and rabbit plasma supplemented with bovine Protein S was sensitive to the anticoagulant activity of activated Protein C. Neither rabbit nor human plasma contained bovine activated Protein C cofactor activity as measured by the enhancement of bovine activated Protein C-catalyzed inactivation of Factor Va. However, bovine activated Protein C was able to inactivate both human and rabbit Factor Va. The inactivation of both of these proteins could be stimulated by the addition of bovine Protein S. These results indicate that the species specificity of bovine activated Protein C is due to the absence of a cofactor protein in non-bovine plasma that will interact with the bovine enzyme. Secondly, these findings further confirm that Protein S is required for the maximal expression of the anticoagulant activity of activated Protein C.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1137-1137
Author(s):  
Mary J. Heeb ◽  
Erning Duan

Abstract Abstract 1137 Background: Platelets contain in their alpha granules ∼2.5% of the protein S in blood. It has been suggested that this protein S supports the anticoagulant activity of exogenous activated protein C (APC), but it is not known whether protein S that is released from stimulated platelets can exert anticoagulant activity that is independent of APC and TFPI. We recently showed that at least some of the anticoagulant activity of plasma protein S is independent of APC and TFPI, although data suggested that plasma protein S may also have TFPI-dependent activity. Objective and methods: To determine if platelet protein S has anticoagulant activity that is independent of APC and TFPI, prothrombinase and extrinsic FXase reactions were initiated on the surface of fresh stimulated or unstimulated washed platelets in the presence and absence of blocking antibodies against APC, TFPI, and/or protein S, or in the presence and absence of purified plasma-derived protein S. Platelets were adjusted to a concentration of 0.7 to 2 × 10e8/ml, which contained 2.3–6.5 nM total platelet protein S. The last platelet wash contained negligible amounts of plasma protein S. Results: Neutralizing anti-protein S antibodies allowed up to 5.7-fold (mean: 2.1 ± 1.5 –fold, n=13) more thrombin generation on calcium ionophore-stimulated platelets following supplementation with 50–500 pM FXa and 600 nM prothrombin, and allowed up to 2.5-fold (mean: 1.7 ± 0.7 –fold, n=11) more thrombin generation on platelets that were not ionophore-stimulated but were gradually stimulated following FXa and prothrombin supplementation. Anti-protein S antibodies had no effect on thrombin generation on platelets that were treated with prostaglandin E1 (PGE1) to suppress platelet activation and then supplemented with procoagulants. This implies that platelet protein S is released from stimulated platelets and downregulates thrombin generation on platelets, and that neutralizing anti-protein S antibodies block this activity of protein S. Anti-protein S antibodies allowed up to 1.8-fold (mean 1.5 ± 0.2 –fold, n=8) more FXa generation on the surface of stimulated platelets supplemented with 5 pM TF, 100 pM FVIIa, and 160 nM FX, but anti-protein S antibodies had no effect on FXa generation on platelets treated with PGE1. Most of these experiments were performed in the presence of neutralizing antibodies against TFPI and APC, but thrombin and FXa generation on platelets under the varying conditions described were unaffected by the presence of these neutralizing antibodies. Purified plasma-derived zinc-containing protein S downregulated thrombin and FXa generation on platelets (IC50 = 6–18 nM PS) and in plasma >10-fold more potently than zinc-deficient protein S. We could not demonstrate a synergistic anticoagulant effect when TFPI was combined with zinc-deficient protein S in the presence of stimulated platelets and procoagulant proteins. Conclusion: Protein S that is released from stimulated platelets exerts anticoagulant activity that is independent of TFPI and APC. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document