Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
2.
Thromb Res ; 202: 84-89, 2021 06.
Article in English | MEDLINE | ID: mdl-33784534

ABSTRACT

INTRODUCTION: Venous thrombosis is the leading cause of pregnancy-related maternal morbidity and mortality. The thrombosis risk is increased by caesarean section and blood loss, though underlying mechanisms of these prothrombotic changes remain unknown. MATERIALS AND METHODS: This prospective study recruited 50 pregnant women at term undergoing elective caesarean section at University Hospital Magdeburg, Germany. Blood loss during surgery was correlated with the changes in total protein S, full length TFPI (TFPIfl), prothrombin, the endogenous thrombin potential (ETP) and resistance to activated protein C (APCsr) determined via calibrated automated thrombography. RESULTS: Mean blood loss was 506 ml (95%CI: 456 to 557 ml). Total protein S was 0.63 (95%CI: 0.60 to 0.67) U/ml preoperatively, decreased by 14.8% after caesarean section and almost normalised five days later. TFPIfl was 0.47 (95%CI: 0.41 to 0.53) U/ml before, remained unchanged immediately after and increased by 11.5% five days after surgery. Prothormbin was 1.10 (95%CI: 1.03 to 1.16) U/ml preoperatively, reduced by 10.4% immediately after and increased again five days after caesarean section, exceeding the preoperative values by 4.4% (-0.7 to 9.6). The ETP decreased by 3.9%, whereas the APCsr increased by 37.0% immediately after caesarean section. The changes in total protein S, prothrombin, thrombin generation and APC resistance showed a trend to be more pronounced in the subgroups with higher blood loss. DISCUSSION: Moderate blood loss during caesarean section hardly reduces thrombin generation but aggravates pregnancy-induced APC resistance and combined deficiency of TFPI and protein S, which can account for the increased thrombosis risk in early puerperium.


Subject(s)
Activated Protein C Resistance , Cesarean Section , Blood Coagulation , Cesarean Section/adverse effects , Female , Germany , Humans , Pregnancy , Prospective Studies
3.
Thromb Res ; 200: 16-22, 2021 04.
Article in English | MEDLINE | ID: mdl-33513452

ABSTRACT

BACKGROUND: Decreased blood coagulation factor (F)XIa levels have been shown to protect from thrombosis without bleeding side effects, but less is known on effects of increased FXIa levels. Studies are hampered by lack of a reliable and robust method for FXIa quantification in blood. We aim to develop a new assay employing a unique multivalent catch-and-release system. The system selectively isolates and protects homodimeric FXIa from plasma and releases free FXIa allowing subsequent quantification. METHODS: A dynamic multivalent construct was synthesized by complexing four identical FXIa inhibitors from the snake Bungarus Fasxiatus to avidin through desthiobiotin-PEG-linkers, allowing dissociation of FXIa by excess biotin. PEG-linker lengths were optimised for FXIa inhibitory activity and analysed by Michaelis-Menten kinetics. Finally, the catch-and-release assay was validated in buffer and plasma model systems. RESULTS: Monovalent and multivalent inhibitor constructs were successfully obtained by total chemical synthesis. Multimerisation of Fasxiator resulted in a 30-fold increase in affinity for FXIa from 1.6 nM to 0.05 nM. With use of this system, FXIa could be quantified down to a concentration of 7 pM in buffer and 20 pM in plasma. CONCLUSION: In this proof-of-concept study, we have shown that the catch-and-release approach is a promising technique to quantify FXIa in plasma or buffer. By binding FXIa to the multivalent construct directly after blood drawing, FXIa is hypothesized to be inaccessible for serpin inhibition or auto inactivation. This results in a close reflection of actual circulating FXIa levels at the moment of blood drawing.


Subject(s)
Factor XIa , Thrombosis , Factor XIa/metabolism , Humans , Kinetics
4.
J Thromb Haemost ; 15(5): 950-960, 2017 05.
Article in English | MEDLINE | ID: mdl-28211163

ABSTRACT

Essentials Protein S is a cofactor of activated protein C (APC) and tissue factor pathway inhibitor (TFPI). There are no assays to quantify separate APC and TFPI cofactor activities of protein S in plasma. We developed assays to measure the APC- and TFPI-cofactor activities of protein S in plasma. The assays were sensitive to protein S deficiency, and not affected by the Factor V Leiden mutation. SUMMARY: Background Protein S plays an important role in the down-regulation of coagulation as cofactor for activated protein C (APC) and tissue factor pathway inhibitor (TFPI). Aim To develop functional assays to quantify the APC- and TFPI-cofactor activities of protein S in plasma. Methods APC- and TFPI-cofactor activities of protein S in plasma were measured using calibrated automated thrombography in protein S-depleted plasma supplemented with a small amount of sample plasma either in the presence of anti-TFPI antibodies and APC (APC-cofactor activity) or at excess full-length TFPI without APC (TFPI-cofactor activity). Total and free protein S levels in plasma were measured by ELISAs. Results Average APC-cofactor activities of protein S were 113%, 108% and 89% in plasma from normal individuals (n = 15), FV Leiden heterozygotes (n = 14) and FV Leiden homozygotes (n = 7), respectively, whereas the average APC-cofactor activity of protein S in plasma from heterozygous protein S-deficient individuals (n = 21) was significantly lower (55%). Similar trends were observed for the TFPI-cofactor activity of protein S, with averages of 109%, 115% and 124% in plasma from individuals with normal protein S levels and different FV Leiden genotypes, and 64% in plasma from protein S-deficient patients. APC-cofactor activities of protein S correlated significantly with free and total protein S antigen levels, whereas TFPI-cofactor activities correlated less with protein S antigen levels. Conclusion We have developed functional protein S assays that measure both the APC- and TFPI-cofactor activities of protein S in plasma, which are hardly if at all affected by the FV Leiden mutation.


Subject(s)
Blood Coagulation Tests/methods , Blood Coagulation , Lipoproteins/blood , Protein C/metabolism , Protein S Deficiency/diagnosis , Protein S/metabolism , Thrombin/metabolism , Activated Protein C Resistance/blood , Activated Protein C Resistance/diagnosis , Activated Protein C Resistance/genetics , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Factor V/genetics , Humans , Point Mutation , Predictive Value of Tests , Protein S/genetics , Protein S Deficiency/blood , Protein S Deficiency/genetics
5.
J Thromb Haemost ; 15(1): 140-149, 2017 01.
Article in English | MEDLINE | ID: mdl-27801970

ABSTRACT

Essentials The C-terminus of tissue factor pathway inhibitor (TFPIα) binds to the B-domain of factor V (FV). The functional consequences of this interaction were investigated in plasma and model systems. The TFPIα C-terminus inhibited thrombin generation in plasma, but not in the presence of FVa. The TFPIα C-terminus inhibited FV activation by preventing cleavage at Arg1545 . SUMMARY: Background Factor V (FV) is a carrier and a cofactor of the anticoagulant protein tissue factor pathway inhibitor-α (TFPIα), whose basic C-terminus binds to an acidic region in the B-domain of FV. Proteolysis of FV at Arg709 , Arg1018 and Arg1545 by activated FX (FXa) or thrombin removes the B-domain, and converts FV into a procoagulant cofactor (activated FV [FVa]) of FXa in the prothrombinase complex. However, retention of the acidic region in partially activated FV makes prothrombinase activity susceptible to inhibition by TFPIα. Objective/Methods To investigate the effect of the TFPIα C-terminal peptide (TFPIα C-term) on thrombin generation in plasma and on FV activation in model systems. Results TFPIα C-term inhibited tissue factor-initiated and FXa-initiated thrombin generation in a dose-dependent manner. Failure to inhibit thrombin generation in FV-depleted plasma reconstituted with FVa indicated that the peptide effect was mediated by the acidic region of FV, and was localized at the level of FV activation and/or prothrombinase. In model systems, TFPIα C-term inhibited both FV activation and prothrombinase activity. Western blot analysis showed that the peptide impaired cleavage at Arg1545 by both thrombin and FXa. The inhibition was stronger for FV-short, which binds TFPIα with higher affinity. Similar results were obtained with full-length TFPIα. Conclusions Cleavage of FV at Arg1545 , which abolishes the anticoagulant properties of FV and commits FV to the procoagulant pathway, is inhibited by binding of the TFPIα C-terminus to the FV acidic region. Possible targets of this new anticoagulant function of TFPIα are low-abundance FV(a) species retaining the acidic region.


Subject(s)
Factor V/chemistry , Lipoproteins/chemistry , Adult , Anticoagulants/chemistry , Arginine/chemistry , Binding Sites , Female , Healthy Volunteers , Humans , Male , Peptides/chemistry , Protein Binding , Protein Domains , Thrombin/chemistry
6.
J Thromb Haemost ; 13(1): 92-100, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25348176

ABSTRACT

BACKGROUND: Tissue factor pathway inhibitor-α (TFPIα) inhibits factor Xa by forming a binary TFPI-FXa complex in a reaction that is stimulated by protein S. TF-FVIIa forms a quaternary complex with TFPIα and FXa, which shuts off the initiation of coagulation via the extrinsic pathway. AIM: To investigate whether direct inhibition of FXa by TFPIα independently of TF plays a role in downregulating coagulation. METHODS: Inhibition of FXa by TFPIα in plasma was determined by measuring thrombin generation triggered with FXa, the FX activator from Russell's viper venom (RVV-X), FXIa, or FIXa. TF-independent anticoagulant activities of TFPIα and its cofactor, protein S, were quantified: (i) after neutralization of TFPIα and protein S with anti-TFPI or anti-protein S antibodies; and (ii) in TFPI-depleted or protein S-depleted plasmas supplemented with varying amounts of TFPIα or protein S. RESULTS: Both anti-TFPI and anti-protein S antibodies enhanced thrombin generation in plasma triggered with RVV-X, FXa, FIXa, or FXIa. Anti-TFPI and anti-protein S antibodies decreased the lag time and increased the peak height of thrombin generation to the same extent, indicating that inhibition of FXa by TFPIα requires the presence of protein S. TFPIα and protein S titrations in TFPI-depleted or protein S-depleted plasma in which thrombin formation was initiated with triggers other than TF also revealed TF-independent anticoagulant activity of TFPIα, which was completely dependent on the presence of protein S. CONCLUSION: Direct inhibition of FXa by TFPIα contributes to the downregulation of coagulation.


Subject(s)
Blood Coagulation , Lipoproteins/metabolism , Thrombin/metabolism , Thromboplastin/metabolism , Blood Coagulation Tests , Down-Regulation , Factor IXa/metabolism , Factor Xa/metabolism , Humans , Kinetics , Protein S/metabolism
7.
J Thromb Haemost ; 12(11): 1826-37, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25163770

ABSTRACT

BACKGROUND: TFPI is a Kunitz-type protease inhibitor that downregulates the extrinsic coagulation pathway by inhibiting factor Xa (FXa) and FVIIa. All three Kunitz domains (KD1, KD2, and KD3) and protein S are required for optimal inhibition of FXa and FVIIa. There is limited information on Kunitz domain requirements of the inhibition of TF:FVIIa-catalyzed FIX and FX activation by TFPI. AIM: To investigate the role of the Kunitz domains of TFPI and protein S in the inhibition of FX and FIX activation. METHODS: Inhibition of TF:FVIIa-catalyzed FX and FIX activation by full-length TFPI (TFPIFL ) and TFPI constructs was quantified from progress curves of FXa and FIXa generation measured with chromogenic substrates. RESULTS AND CONCLUSIONS: TFPIFL inhibited TF:FVIIa-catalyzed FIX activation with a Ki of 16.7 nmol L(-1) . Protein S reduced the Ki to 1.0 nmol L(-1) . TFPI1-150 and KD1-KD2 had 10-fold higher Ki values and were not stimulated by protein S. Single Kunitz domains were poor inhibitors of TF:FVIIa-catalyzed FIX activation (Ki >800 nm). FX activation was measured at limiting FVIIa and excess TF or vice versa. At both conditions, TFPIFL , TFPI1-150 , and KD1-KD2 showed similar inhibition of FX activation. However, at low phospholipid concentrations, TFPIFL was ~ 15-fold more active than TFPI1-150 or KD1-KD2. Apparently, excess phospholipids act as a kind of sink for TFPIFL , limiting its availability for TF:FVIIa inhibition. Preformed FXa:TFPIFL/1-150 complexes rapidly and stoichiometrically inhibited FIX and FX activation by TF:FVIIa, indicating that binary TFPI:FXa complex formation is the limiting step in TF:FVIIa inhibition. Protein S also enhanced inhibition of TF:FVIIa-catalyzed FX activation by TFPI.


Subject(s)
Blood Coagulation , Factor IXa/metabolism , Factor VIIa/metabolism , Factor Xa Inhibitors/metabolism , Factor Xa/metabolism , Lipoproteins/metabolism , Thromboplastin/metabolism , Blood Coagulation/drug effects , Catalysis , Dose-Response Relationship, Drug , Factor VIIa/antagonists & inhibitors , Factor Xa Inhibitors/pharmacology , Humans , Kinetics , Lipoproteins/pharmacology , Phospholipids/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein S/metabolism , Recombinant Proteins/metabolism
8.
J Thromb Haemost ; 11(4): 704-14, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23347185

ABSTRACT

BACKGROUND: Tissue factor pathway inhibitor (TFPI) is a multi-Kunitz domain protease inhibitor that down-regulates the extrinsic coagulation pathway by inhibiting FXa and FVIIa. OBJECTIVES: To investigate the role of the three Kunitz domains (KDs) of TFPI in FVIIa inhibition using full-length TFPI (TFPIfl ) and truncated TFPI constructs. METHODS: Inhibition of FVIIa with/without relipidated tissue factor (TF) or soluble TF (sTF) by TFPIfl /TFPI constructs was quantified with a FVIIa-specific chromogenic substrate. RESULTS AND CONCLUSIONS: TFPIfl inhibited TF-FVIIa via a monophasic reaction, which is rather slow at low TFPIfl concentrations (t½  ≈ 5 min at 2 nm TFPI) and has a Ki of 4.6 nm. In the presence of sTF and without TF, TFPIfl was a poor FVIIa inhibitor, with Ki values of 122 nm and 1118 nm, respectively. This indicates that phospholipids and TF significantly contribute to FVIIa inhibition by TFPIfl . TFPI constructs without the KD3-c-terminus (TFPI1-150 and KD1-KD2) were 7-10-fold less effective than TFPIfl in inhibiting TF-FVIIa and sTF-FVIIa, indicating that the KD3-C-terminus significantly contributes to direct inhibition of FVIIa by TFPI. Compared with KD1-KD2, KD1 was a poor TF-FVIIa inhibitor (Ki =434 nm), which shows that the KD2 domain of TFPI also contributes to FVIIa inhibition. Protein S stimulated TF-FVIIa inhibition by TFPIfl (Ki =0.7 nm). In the presence of FXa, a tight quaternary TF-FVIIa-TFPI-FXa complex is formed with TFPIfl , TFPI1-150 and KD1-KD2, with Ki values of < 0.15 nm, 0.5 nm and 0.8 nm, respectively, indicating the KD3-C-terminus is not a prerequisite for quaternary complex formation. Phospholipids and the Gla-domain of FXa are required for quaternary complex formation.


Subject(s)
Factor VIIa/antagonists & inhibitors , Lipoproteins/pharmacology , Amino Acid Sequence , Humans , Lipoproteins/chemistry , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
10.
Br J Pharmacol ; 166(6): 1833-45, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22324472

ABSTRACT

BACKGROUND AND PURPOSE Endothelin-1 (ET-1) causes long-lasting vasoconstrictions. These can be prevented by ET(A) receptor antagonists but are only poorly reversed by these drugs. We tested the hypothesis that endothelin ET(A) receptors are susceptible to allosteric modulation by endogenous agonists and exogenous ligands. EXPERIMENTAL APPROACH Rat isolated mesenteric resistance arteries were pretreated with capsaicin and studied in wire myographs, in the presence of L-NAME and indomethacin to concentrate on arterial smooth muscle responses. KEY RESULTS Endothelins caused contractions with equal maximum but differing potency (ET-1 = ET-2 > ET-3). ET-1(1-15) neither mimicked nor antagonized these effects in the absence and presence of ET(16-21). 4(Ala) ET-1 (ET(B) agonist) and BQ788 (ET(B) antagonist) were without effects. BQ123 (peptide ET(A) antagonist) reduced the sensitivity and relaxed the contractile responses to endothelins. Both effects depended on the agonist (pK(B): ET-3 = ET-1 > ET-2; % relaxation: ET-3 = ET-2 > ET-1). Also, with PD156707 (non-peptide ET(A) antagonist) agonist-dependence and a discrepancy between preventive and inhibitory effects were observed. The latter was even more marked with bulky analogues of BQ123 and PD156707. CONCLUSIONS AND IMPLICATIONS These findings indicate allosteric modulation of arterial smooth muscle ET(A) receptor function by endogenous agonists and by exogenous endothelin receptor antagonists. This may have consequences for the diagnosis and pharmacotherapy of diseases involving endothelins.


Subject(s)
Mesenteric Arteries/physiology , Muscle, Smooth, Vascular/physiology , Receptor, Endothelin A/physiology , Animals , Binding, Competitive , Carbocyanines/pharmacology , Dioxoles/pharmacology , Endothelin A Receptor Antagonists , Endothelins/pharmacology , Fluorescein-5-isothiocyanate/pharmacology , In Vitro Techniques , Mesenteric Arteries/drug effects , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Peptides, Cyclic/pharmacology , Rats , Receptor, Endothelin A/agonists
11.
Neurology ; 78(7): 493-8, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22302552

ABSTRACT

OBJECTIVES: New insights in the pathophysiology of lacunar stroke (LS) suggest that it is caused by increased permeability of the blood-brain barrier due to endothelial activation. Because endothelial cells are the major production and storage site of tissue factor pathway inhibitor (TFPI), this protein can be used as marker of endothelial activation. In this observational study we measured the different pools of TFPI, as a marker of endothelial function, in first-ever lacunar stroke patients. METHODS: We determined antigen levels of total and free full-length (FL) TFPI using ELISA in 149 patients and 42 controls. Heparin-releasable free FL TFPI was determined in a random subset of 17 patients and 15 controls. By brain MRI, we classified LS patients as having isolated lacunar infarct (ILA) or silent ischemic lesions (SILs). RESULTS: Plasma levels of total TFPI were highest in patients with SILs compared with those with ILA, but this association disappeared after correction for age and levels of low-density lipoprotein cholesterol. However, levels of heparin-releasable free FL TFPI were higher in patients than in controls. CONCLUSIONS: Although ambient plasma levels of total TFPI were not different in subtypes of LS, the increased levels of heparin-releasable TFPI in patients suggest a role of endothelial activation in the pathogenesis of LS.


Subject(s)
Anticoagulants/pharmacology , Heparin/pharmacology , Lipoproteins/metabolism , Stroke, Lacunar/metabolism , Age Factors , Aged , Biomarkers , Brain Ischemia/metabolism , Brain Ischemia/pathology , Clinical Protocols , Denmark , Endothelium, Vascular/physiology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Image Processing, Computer-Assisted , Logistic Models , Magnetic Resonance Imaging , Male , Middle Aged , Prospective Studies , Registries , Risk Factors , Stroke, Lacunar/classification
12.
J Thromb Haemost ; 9(11): 2243-50, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21895962

ABSTRACT

BACKGROUND: The tissue factor pathway inhibitor (TFPI)/protein S anticoagulant system is a potent inhibitor of blood coagulation. TFPI and protein S are major determinants of thrombin generation (TG) tests determined at low tissue factor (TF) and at high TF concentrations in the presence of activated protein C (APC). Both TFPI and protein S protect against venous thrombosis, but the importance of the TFPI/protein S system in arterial thrombosis remains unclear. OBJECTIVES: To investigate the influence of the TFPI/protein S anticoagulant system on the risk of myocardial infarction (MI) in young women. METHODS: The RATIO study is a case-control study in women under 50 years of age, including 205 patients and 638 controls. TFPI and protein S were quantified using ELISA. The TFPI/protein S activity (nTFPIr) and the APC sensitivity ratio (nAPCsr) were determined using TG tests. Odds ratios (ORs) adjusted for putative confounders and corresponding 95% confidence intervals (95% CI) were determined. RESULTS: Women with MI had higher TFPI levels than controls (135.9 ± 40% vs. 124.2 ± 41%), resulting in increased TFPI/protein S activities and increased APC sensitivity. Furthermore, an increased TFPI activity was associated with MI [nTFPIr: adjusted OR Q1 vs. Q4 = 2.1 (95%CI 1.1-4.1)]. Additionally, an increased APC sensitivity was associated with MI [nAPCsr: adjusted OR Q1 vs. Q4 = 1.7 (95% CI 0.9-3.2)] CONCLUSION: Women with MI had increased TFPI levels compared with controls. Consequently, the TFPI/protein S activity and APC sensitivity are increased in women with MI. Whether this increase in TFPI activity acts as a compensating mechanism for an increased procoagulant state or is a marker of endothelial damage remains to be investigated.


Subject(s)
Lipoproteins/metabolism , Myocardial Infarction/blood , Adult , Anticoagulants , Cardiotonic Agents , Case-Control Studies , Female , Humans , Lipoproteins/blood , Middle Aged , Odds Ratio , Protein S/analysis , Up-Regulation , Young Adult
13.
J Thromb Haemost ; 9(1): 209-19, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20961394

ABSTRACT

BACKGROUND AND OBJECTIVES: The non-allelic variant of CXCL4/PF4, CXCL4L1/PF4alt, differs from CXCL4 in three amino acids of the C-terminal α-helix and has been characterized as a potent anti-angiogenic regulator. Although CXCL4 structurally belongs to the chemokine family, it does not behave like a 'classical' chemokine, lacking significant chemotactic properties. Specific hallmarks are its angiostatic, anti-proliferative activities, and proinflammatory functions, which can be conferred by heteromer-formation with CCL5/RANTES enhancing monocyte recruitment. METHODS AND RESULTS: Here we show that tube formation of endothelial cells was inhibited by CXCL4L1 and CXCL4, while only CXCL4L1 triggered chemokinesis of endothelial cells. The chemotactic response towards VEGF and bFGF was attenuated by both variants and CXCL4L1-induced chemokinesis was blocked by bFGF or VEGF. Endothelial cell proliferation was inhibited by CXCL4 (IC(50) 6.9 µg mL(-1)) but not by CXCL4L1, while both chemokines bound directly to VEGF and bFGF. Moreover, CXCL4 enhanced CCL5-induced monocyte arrest in flow adhesion experiments and monocyte recruitment into the mouse peritoneal cavity in vivo, whereas CXCL4L1 had no effect. CXCL4L1 revealed lower affinity to CCL5 than CXCL4, as quantified by isothermal fluorescence titration. As evidenced by the reduction of the activated partial thromboplastin time, CXCL4L1 showed a tendency towards less heparin-neutralizing activity than CXCL4 (IC(50) 2.45 vs 0.98 µg mL(-1)). CONCLUSIONS: CXCL4L1 may act angiostatically by causing random endothelial cell locomotion, disturbing directed migration towards angiogenic chemokines, serving as a homeostatic chemokine with a moderate structural distinction yet different functional profile from CXCL4.


Subject(s)
Angiostatic Proteins/metabolism , Cell Movement , Cell Proliferation , Chemotaxis, Leukocyte , Endothelial Cells/immunology , Inflammation Mediators/metabolism , Monocytes/immunology , Neovascularization, Physiologic , Platelet Factor 4/metabolism , Angiostatic Proteins/chemical synthesis , Angiostatic Proteins/genetics , Animals , Blood Coagulation , Cells, Cultured , Chemokine CCL5/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Inflammation Mediators/chemical synthesis , Mice , Mice, Inbred C57BL , Partial Thromboplastin Time , Platelet Factor 4/chemical synthesis , Platelet Factor 4/genetics , Protein Multimerization , Recombinant Proteins/metabolism , Time Factors , Vascular Endothelial Growth Factor A/metabolism
14.
J Thromb Haemost ; 8(4): 750-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20088940

ABSTRACT

BACKGROUND: Protein S acts as a cofactor for full-length tissue factor pathway inhibitor (TFPI) in the downregulation of thrombin formation. OBJECTIVE: To develop a functional test to measure the activity of the TFPI-protein S system in plasma. METHODS/PATIENTS: Using calibrated automated thrombography, we quantified the activity of the TFPI-protein S system in plasma by measuring thrombin generation in the absence and presence of neutralizing antibodies against protein S or TFPI. Moreover, we designed an enzyme-linked immunosorbent assay (ELISA) to determine the level of full-length TFPI in plasma. The performance of these assays was examined in plasma from 85 normal individuals and from 35 members of protein S-deficient families. RESULTS: The ratio of thrombin peaks determined in the absence and presence of anti-protein S antibodies (protein S ratio = 0.5 in normal plasma) is a measure of the TFPI cofactor activity of protein S, whereas the ratio of thrombin peaks determined in the absence and presence of anti-TFPI antibodies (TFPI ratio = 0.25 in normal plasma) is a measure of the overall activity of the TFPI-protein S system. Protein S and TFPI ratios were elevated in protein S-deficient individuals, indicating an impairment of the TFPI-protein S system. Both ratios correlated well with full-length TFPI levels, which were significantly lower in protein S-deficient patients than in normal family members. CONCLUSIONS: Functional assays for the TFPI-protein S system and an ELISA for full-length TFPI were developed. These assays show that the activity of the TFPI-protein S anticoagulant pathway is impaired in individuals with congenital protein S deficiency.


Subject(s)
Blood Coagulation Tests , Enzyme-Linked Immunosorbent Assay , Lipoproteins/blood , Protein S Deficiency/diagnosis , Protein S/metabolism , Thrombin/metabolism , Amino Acid Sequence , Antibodies, Neutralizing , Automation, Laboratory , Biomarkers/blood , Blood Coagulation Tests/standards , Calibration , Case-Control Studies , Enzyme-Linked Immunosorbent Assay/standards , Female , Humans , Lipoproteins/immunology , Male , Molecular Sequence Data , Predictive Value of Tests , Protein S/immunology , Protein S Deficiency/blood , Reproducibility of Results
15.
J Thromb Haemost ; 8(2): 294-300, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20002538

ABSTRACT

BACKGROUND: Protein S and tissue factor pathway inhibitor (TFPI) act together in down-regulating coagulation. OBJECTIVE: To investigate the TFPI/protein S system in hereditary and acquired protein S deficiency. METHODS: Plasma antigen levels of protein S and full-length TFPI were determined in heterozygous type I protein S-deficient individuals (n=35), patients on oral anticoagulant treatment (OAT) (n=29), oral contraceptive (OC) users (n=10) and matched controls. Thrombin generation was determined using calibrated automated thrombography. RESULTS: Full-length TFPI levels were lower in type I protein S-deficient individuals (76.8+/-33.8%) than in age- and sex-matched controls (128.0+/-59.4%, P<0.001). Among protein S-deficient individuals with thrombosis, those on OAT had not only lower total protein S levels (25.7+/-8.2% vs. 54.7+/-8.2%, P<0.001), but also lower full-length TFPI levels (52.6+/-15.0% vs. 75.4+/-22.9%, P=0.009) than those not on OAT. Similarly, OC users had lower protein S (73.8+/-11.5% vs. 87.9+/-10.8%, P=0.005) and full-length TFPI levels (73.7+/-27.7% vs. 106.4+/-29.2%, P=0.007) than non-users. When triggered with tissue factor, plasma from protein S-deficient individuals generated 3-5-fold more thrombin than control plasma. The difference was only partially corrected by normalization of the protein S level, full correction requiring additional normalization of the TFPI level. Protein S-immunodepletion experiments indicated that free protein S and full-length TFPI form a complex in plasma, and the protein S/TFPI interaction was confirmed by surface plasmon resonance analysis. CONCLUSIONS: Full-length TFPI binds to protein S in plasma and is reduced in genetic and acquired protein S deficiency. The concomitant TFPI deficiency substantially contributes to the hypercoagulable state associated with protein S deficiency.


Subject(s)
Blood Coagulation , Lipoproteins/blood , Protein S Deficiency/blood , Protein S/metabolism , Administration, Oral , Adult , Aged , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Blood Coagulation/drug effects , Blood Coagulation/genetics , Case-Control Studies , Contraceptives, Oral/adverse effects , Down-Regulation , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Male , Middle Aged , Protein Binding , Protein S/genetics , Protein S Deficiency/chemically induced , Protein S Deficiency/genetics , Risk Factors , Thrombin/metabolism , Thromboplastin/metabolism , Time Factors , Young Adult
16.
J Thromb Haemost ; 7 Suppl 1: 165-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19630792

ABSTRACT

Protein S is an anticoagulant cofactor of full-length tissue factor pathway inhibitor (TFPI) that facilitates optimal factor Xa-inhibition and efficient down-regulation of thrombin generation in plasma. Protein S and TFPI are constitutively active in plasma and therefore provide an effective anticoagulant barrier against unwanted procoagulant activity in the circulation. In this review, we describe the current status on how TFPI-activity depends on protein S, and show that TFPI and protein S are major regulators of thrombin generation both in the absence and presence of activated protein C (APC). As there is covariation of plasma TFPI and protein S levels both in health and in disease, these findings suggest that the risk of venous thrombosis associated with protein S deficiency states might be in part explained by the accompanying low plasma TFPI levels.


Subject(s)
Lipoproteins/physiology , Protein S/physiology , Humans , Thrombin/biosynthesis , Venous Thrombosis/etiology
17.
J Thromb Haemost ; 6(12): 2132-42, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18826391

ABSTRACT

BACKGROUND: Collagen acts as a potent surface for platelet adhesion and thrombus formation under conditions of blood flow. Studies using collagen-derived triple-helical peptides have identified the GXX'GER motif as an adhesive ligand for platelet integrin alpha2beta1, and (GPO)(n) as a binding sequence for the signaling collagen receptor, glycoprotein VI (GPVI). OBJECTIVE: The potency was investigated of triple-helical peptides, consisting of GXX'GER sequences within (GPO)(n) or (GPP)(n) motifs, to support flow-dependent thrombus formation. RESULTS: At a high-shear rate, immobilized peptides containing both the high-affinity alpha2beta1-binding motif GFOGER and the (GPO)(n) motif supported platelet aggregation and procoagulant activity, even in the absence of von Willebrand factor (VWF). With peptides containing only one of these motifs, co-immobilized VWF was needed for thrombus formation. The (GPO)(n) but not the (GPP)(n) sequence induced GPVI-dependent platelet aggregation and procoagulant activity. Peptides with intermediate affinity (GLSGER, GMOGER) or low-affinity (GASGER, GAOGER) alpha2beta1-binding motifs formed procoagulant thrombi only if both (GPO)(n) and VWF were present. At a low-shear rate, immobilized peptides with high- or low-affinity alpha2beta1-binding motifs mediated formation of thrombi with procoagulant platelets only in combination with (GPO)(n). CONCLUSIONS: Triple-helical peptides with specific receptor-binding motifs mimic the properties of native collagen I in thrombus formation by binding to both platelet collagen receptors. At a high-shear rate, either GPIb or high-affinity (but not low-affinity) GXX'GER mediates GPVI-dependent formation of procoagulant thrombi. By extension, high-affinity binding for alpha2beta1 can control the overall platelet-adhesive activity of native collagens.


Subject(s)
Collagen/chemistry , Integrin alpha2beta1/metabolism , Peptide Fragments/metabolism , Platelet Adhesiveness , Platelet Membrane Glycoproteins/metabolism , Thrombosis/etiology , Amino Acid Motifs , Amino Acid Sequence , Cells, Cultured , Humans , Molecular Mimicry , Peptide Fragments/chemical synthesis , Protein Binding , von Willebrand Factor/metabolism
18.
Thromb Res ; 122 Suppl 1: S60-3, 2008.
Article in English | MEDLINE | ID: mdl-18691502

ABSTRACT

Protein S is a vitamin K-dependent protein that acts as a cofactor of the anticoagulant protein APC. However, protein S also exhibits anticoagulant activity in the absence of APC. Thrombin generation experiments in normal plasma and in plasma deficient in tissue factor pathway inhibitor (TFPI) and/or protein S demonstrated that protein S stimulates the inhibition of TF by TFPI. Kinetic analysis in model systems containing purified proteins showed that protein S enhances the formation of the binary FXa:TFPI complex by reducing the Ki of TFPI from approximately 4 nM to approximately 0.5 nM. Enhancement of inhibitory activity of TFPI by protein S is only observed with full-length TFPI and in the presence of a negatively charged phospholipid surface. The Ki decrease brings the TFPI concentration necessary for FXa:TFPI complex formation within range of the plasma TFPI concentration which increases FXa:TFPI complex formation and accelerates feedback inhibition of the TF pathway by enhancing the formation of the quaternary TFPI:FXa:TF:FVIIa complex. Thus, protein S is not only a cofactor of APC, but also of TFPI. A reduced TFPI cofactor activity may contribute to the increased risk of venous thrombosis in protein-S deficient individuals. Using calibrated automated thrombography we have developed two assays that enable quantification of the functional activity of the TFPI/protein S system in plasma. These assays show that the activity of the TFPI/protein S system is greatly impaired in oral contraceptive users.


Subject(s)
Lipoproteins/metabolism , Protein S/metabolism , Thromboplastin/antagonists & inhibitors , Thromboplastin/metabolism , Blood Coagulation Factors/metabolism , Factor Xa Inhibitors , Humans , Kinetics , Lipoproteins/analysis , Lipoproteins/genetics , Protein S/analysis , Protein S/pharmacology , Receptors, Cell Surface/metabolism , Thrombin/biosynthesis , Thrombosis/pathology
19.
J Thromb Haemost ; 5(12): 2503-11, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17848178

ABSTRACT

BACKGROUND: Matrix Gla protein (MGP) is a small vitamin K-dependent protein containing five gamma-carboxyglutamic acid (Gla) residues that are believed to be important in binding Ca(2+), calcium crystals and bone morphogenetic protein. In addition, MGP contains phosphorylated serine residues that may further regulate its activity. In vivo, MGP has been shown to be a potent inhibitor of vascular calcification; however, the precise molecular mechanism underlying the function of MGP is not yet fully understood. METHODS AND RESULTS: We investigated the effects of MGP in human vascular smooth muscle cell (VSMC) monolayers that undergo calcification after exposure to an increase in Ca(2+) concentration. Increased calcium salt deposition was found in cells treated with the vitamin K antagonist warfarin as compared to controls, whereas cells treated with vitamin K(1) showed decreased calcification as compared to controls. With conformation-specific antibodies, it was confirmed that warfarin treatment of VSMCs resulted in uncarboxylated (Gla-deficient) MGP. To specifically test the effects of MGP on VSMC calcification, we used full-length synthetic MGP and MGP-derived peptides representing various domains in MGP. Full length MGP, the gamma-carboxylated motif (Gla) (amino acids 35-54) and the phosphorylated serine motif (amino acids 3-15) inhibited calcification. Furthermore, we showed that the peptides were not taken up by VSMCs but bound to the cell surface and to vesicle-like structures. CONCLUSIONS: These data demonstrate that both gamma-glutamyl carboxylation and serine phosphorylation of MGP contribute to its function as a calcification inhibitor and that MGP may inhibit calcification via binding to VSMC-derived vesicles.


Subject(s)
Calcinosis/prevention & control , Calcium-Binding Proteins/metabolism , Calcium/metabolism , Extracellular Matrix Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Processing, Post-Translational , 1-Carboxyglutamic Acid/metabolism , Adolescent , Adult , Amino Acid Sequence , Calcinosis/metabolism , Calcium-Binding Proteins/chemistry , Cell Membrane/metabolism , Cells, Cultured , Extracellular Matrix Proteins/chemistry , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Peptide Fragments/metabolism , Phosphorylation , Protein Structure, Tertiary , Serine/metabolism , Transport Vesicles/metabolism , Vitamin K/antagonists & inhibitors , Vitamin K/metabolism , Vitamin K 1/pharmacology , Warfarin/pharmacology , Matrix Gla Protein
20.
J Thromb Haemost ; 2(10): 1766-73, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15456488

ABSTRACT

BACKGROUND: Plasma protein S normally circulates free (40%) or complexed with C4b-binding protein (PS-C4BP); only free protein S is a cofactor for activated protein C during factor (F) Va inactivation. Protein S-Heerlen lacks a carbohydrate group, leading to low plasma free protein S levels, but normal levels of PS-C4BP. OBJECTIVES: Because protein S-Heerlen is not associated with thrombosis, we investigated whether PS-C4BP is directly anticoagulant in plasma and whether PS-Heerlen-C4BP has enhanced direct anticoagulant activity. METHODS: An assay for protein S direct activity was applied to Heerlen-heterozygous plasmas. Free and complexed protein S were repeatedly isolated from normal and Heerlen-heterozygous plasmas and tested for direct anticoagulant activity in prothrombinase assays and in plasma. RESULTS: Heerlen-heterozygous plasmas were deficient in free and total protein S antigen but had normal to high protein S direct anticoagulant activity. Purified Heerlen-heterozygous PS-C4BP was 7-fold more potent than normal PS-C4BP in inhibiting full prothrombinase activity, and 22-fold more potent in inhibiting prothrombin activation in the absence of FVa; it also specifically prolonged plasma clotting times 14-fold more than normal PS-C4BP. Heerlen-heterozygous PS-C4BP did not compete for limiting phospholipids any better than normal PS-C4BP. However, ligand blots and surface plasmon resonance studies showed that Heerlen-heterozygous PS-C4BP bound more avidly to FXa than did normal PS-C4BP (apparent Kd = 4.3 nm vs. 82 nm). CONCLUSIONS: Plasma-derived PS-C4BP has direct anticoagulant activity in plasma and in purified systems. Enhanced direct activity of PS-Heerlen-C4BP may compensate for low free protein S levels and low cofactor activity in individuals with protein S-Heerlen.


Subject(s)
Histocompatibility Antigens/metabolism , Protein S/analysis , Adult , Anticoagulants , Blood Coagulation Tests , Blood Platelets , Complement C4b-Binding Protein , Factor Xa/metabolism , Family Health , Female , Heterozygote , Histocompatibility Antigens/physiology , Humans , Male , Phospholipids , Protein Binding/genetics , Protein S/metabolism , Protein S/physiology , Prothrombin/antagonists & inhibitors , Thromboplastin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL