Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
1.
Biosens Bioelectron ; 104: 15-20, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29294406

ABSTRACT

The study focuses the dose administration of dabigatran to avoid the deaths due to hemorrhagic complications and thromboembolic stroke in clinics worldwide. To target the issue, a novel emerging acoustic technology, namely ''Quartz Crystal Microbalance with Dissipation'' (QCM-D) has been applied, while the acoustic assays namely ''activated Partial Thromboplastin Time'' (aPTT) and ''Prothrombinase complex-induced Clotting Test'' (PiCT) have been compared with the standard methods in parallel. Both techniques have been applied to 300 samples, including 220 plasma samples of patients suffering coagulation disorders and 80 plasma samples of non-patients. In comparison, the coagulation times of the acoustic aPTT and PiCT yielded an excellent correlation with the standard methods with in analytical standard deviation limits. Finally, the acoustic aPTT assay is the ''gold standard'' for a dose administration of the new oral anticoagulant, where the Δf/ΔΓ ratio of the acoustic assay demonstrates that dabigatran with FEIBA 50 combination could be a safe remedy to avoid the deaths in clinics.


Subject(s)
Biosensing Techniques , Blood Coagulation Disorders/blood , Quartz Crystal Microbalance Techniques , Thrombin/isolation & purification , Blood Coagulation/drug effects , Blood Coagulation Disorders/pathology , Blood Coagulation Tests , Dabigatran/administration & dosage , Factor V/isolation & purification , Factor Xa/isolation & purification , Female , Humans , Male , Partial Thromboplastin Time
3.
Klin Lab Diagn ; (12): 26, 39-41, 2013 Dec.
Article in Russian | MEDLINE | ID: mdl-24757862

ABSTRACT

The article deals with comparing technique of detection of Leiden mutation on the basis of PEXT-reaction with subsequent bioluminescent microanalysis of products with technique based on RT-PCR. The sampling for testing comprised 83 specimen of genome DNA including 35 specimens with known Leiden heterozygote mutation. The commercial kit "SNP-express-PB" (Litex) was used as a comparison test. It is demonstrated that proposed approach is a simple in its application, effective and relatively inexpensive technique of detection of Leiden one-nucleotide polymorphism in gene V of blood coagulation factor. The technique "PED-Biolum" has no differences in comparison with commercial technique RT-PCR concerning ability to detect mutant allele and matches it in parameters of economic effectiveness.


Subject(s)
Factor V/isolation & purification , Luminescent Measurements/methods , Mutation/genetics , Alleles , DNA Primers/chemistry , DNA Primers/genetics , Factor V/genetics , Humans , Polymorphism, Single Nucleotide
4.
Methods Mol Biol ; 885: 71-89, 2012.
Article in English | MEDLINE | ID: mdl-22565991

ABSTRACT

Microvascular thrombosis, following the activation of clotting cascade, is a hallmark of porcine solid organ xenograft rejection. The analysis of differences between human, monkey, and pig coagulation systems is crucial when monkey is used as animal model and pig as organ donor in xenotransplantation. Thrombosis, according to many authors, may be due to the molecular incompatibilities between natural anticoagulants present on pig endothelium and primate activated coagulation factors. The generation of activated protein C (PC) is critical for the physiological anticoagulation. One of the major incompatibilities may be related to the inability of pig thrombomodulin (TM) and endothelial protein C receptor to activate the recipient (primate) circulating PC in the presence of thrombin. Tissue factor pathway inhibitor (TFPI), is the primary inhibitor of tissue factor (TF)-induced coagulation. TFPI directly inhibits the activated factor X (FXa) and blocks the procoagulant activity of the TF/factor VIIa (FVIIa) complex by forming a quaternary TF/FVIIa/FXa/TFPI complex. Microvascular thrombosis, observed in the organ transplant, may also be due to the failure of pig TFPI to bind human FXa efficiently and inhibit human FVIIa/TF activity. The methods described in this chapter can be useful for the identification and characterization of primate and pig coagulation factors (isolated from a small volume of blood) by using SDS-PAGE and immunoblotting. Differences in molecular weight can help in the identification of the origin (pig or primate) of coagulation proteins in plasma from the recipient of xenografts. On the other hand, in vitro models of PC pathway and TFPI on human umbilical vein endothelial cells (HUVEC) and porcine aortic endothelial cells (PAEC) are described which can be used for studying incompatibilities between primate and pig.


Subject(s)
Blood Coagulation/immunology , Transplantation, Heterologous/immunology , Animals , Blood Coagulation Tests , Blood Platelets/immunology , Blood Platelets/metabolism , Cell Separation , Cells, Cultured , Factor V/immunology , Factor V/isolation & purification , Factor V/metabolism , Flow Cytometry , Human Umbilical Vein Endothelial Cells , Humans , Macaca fascicularis , Microscopy, Fluorescence , Primary Cell Culture , Protein C/immunology , Protein C/metabolism , Protein S/immunology , Protein S/isolation & purification , Protein S/metabolism , Swine , Thromboplastin
5.
Pathol Res Pract ; 207(11): 671-3, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21996318

ABSTRACT

Factor V Leiden (FVL; G1691A) is an autosomal dominant mutation with a high risk for thrombosis. Speculation that founders of FVL lived in the Middle East is supported by a prevalence of FVL that is higher in Arabs residing in Israel, Jordan, Lebanon, and Syria (12-14%) than in other white populations like Europeans (4-5%, up to 15% in the South of Sweden). We sought to verify the appropriate use of skin color as a clinical sign by which Arab individuals in Kuwait are included or excluded from testing for FVL. After institutional approval, 200 healthy Arabs residing in Kuwait consented to participate. Skin type was distinguished for the participants by Fitzpatrick natural skin color classification: 76 (38%) skin type II (white), 96 (48%) Mediterranean skin type IV (brown), and 28 (14%) skin type VI (black). FVL was tested by real-time PCR, and the percentage of carriers was calculated in each group. FVL was positive in 17 (8.5%) of the total subjects: 8 (10.5%) skin type II, 7 (7.3%) skin type IV, and 2 (7.1%) skin type VI. Therefore, FVL shows an even distribution in Arabs, and all Arabs residing in Kuwait should be tested for FVL irrespective of skin color.


Subject(s)
Arabs/genetics , Factor V/genetics , Heterozygote , Skin Pigmentation , Thrombosis/genetics , Arabs/ethnology , Factor V/isolation & purification , Gene Frequency , Genotype , Humans , Kuwait , Mutation , Prevalence , Real-Time Polymerase Chain Reaction , Risk Factors , Skin Pigmentation/genetics , Thrombosis/ethnology
6.
Br J Haematol ; 142(6): 962-70, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18643923

ABSTRACT

The procoagulant activity of factors VIII and V depends on the presence of metal ion(s). We examined the effect of cation-exchange resins with different functional groups on both factors, of which only reaction with iminodiacetate resin resulted in the complete loss of their activity levels in plasma. However, the antigen level of factor VIII was preserved by >95%. This resin reduced divalent cations content present in factor VIII preparations, indicating that it inactivated this factor by direct deprivation of predominant Ca(2+) (>Mn(2+)>>Cu(2+)), rather than adsorption of the factor itself. The antigen level of recombinant factor VIII alone was decreased by >95% by reaction with resin, whilst that complexed with von Willebrand factor was preserved by >95%. Iminodiacetate resin-treated plasma was evaluated by measuring factor VIII and V activity in plasma with various levels of either activity. These were significantly correlated to the values obtained using factor VIII- or V-deficient plasma prepared commercially by immunodepletion. We demonstrated that iminodiacetate resin-induced factors VIII and V inactivation is because of direct deprivation of metal ions, predominantly Ca(2+), which is more essential for the functional structure of their molecules. Furthermore, iminodiacetate resin-treated plasma would be useful as a substrate for measuring the activity of these factors.


Subject(s)
Factor VIII/chemistry , Factor V/chemistry , Blood Coagulation Tests/methods , Calcium/chemistry , Copper/chemistry , Factor V/analysis , Factor V/isolation & purification , Factor VIII/analysis , Factor VIII/isolation & purification , Humans , Imino Acids/chemistry , Immunosorbent Techniques , Ion Exchange Resins , Manganese/chemistry
7.
Adv Exp Med Biol ; 614: 101-7, 2008.
Article in English | MEDLINE | ID: mdl-18290319

ABSTRACT

Factor V (FV) is a coagulant in plasma. The FV molecule consists of a heavy chain and a light chain, and Factor V Leiden (FVL) is mutated FV at a single amino acid in the heavy chain. FVL patients are in a dangerous hyper-coagulation state in their body. Current FVL diagnosis is done by DNA analysis, which is expensive and time consuming. Our group has been developing a real-time, cost effective immuno-optical biosensor for FVL diagnosis. For the sensor development, pure FVL, which is not currently available, is needed. Here, we have attempted FVL purification from FVL patient's plasma. Since plasma contains many proteins and some proteins are structurally homologous to FV, the purification must be done by a very specific method, such as immuno-affinity chromatography. However, an antibody that does not react with FV is not currently available. Because the mutation is in the heavy chain and the amino acid sequence of the light chain of FVL is identical to that of FV, antibodies generated against the light chain of FV were tested for purifying FVL. Plasma was obtained from a homozygous FVL patient. First, the plasma was pretreated by barium citrate and polyethylene glycol 6000, to remove the vitamin K-dependent proteins, alpha globulins, and other smaller than 6 kDa molecular weight proteins. The yield in the process was 54%. Immuno-affinity purification of FVL from patient plasma was then performed using an anti-FV light chain antibody immobilized CNBr-Sepharose, and the purification yield was 25%. In summary, the antibody against the light chain of FV was able to purify the single point mutated form of FV (FVL) from plasma with an overall yield of 14%. The same principle can probably be used for purification of the other single point mutated proteins.


Subject(s)
Factor V/genetics , Factor V/isolation & purification , Homozygote , Point Mutation , Amino Acid Substitution , Case-Control Studies , Chromatography, Affinity , Enzyme-Linked Immunosorbent Assay , Humans
8.
J Biol Chem ; 279(4): 2383-93, 2004 Jan 23.
Article in English | MEDLINE | ID: mdl-14594814

ABSTRACT

Platelet- and plasma-derived factor Va (FVa) serve essential cofactor roles in prothrombinase-catalyzed thrombin generation. Platelet-derived FV/Va, purified from Triton X-100 platelet lysates was composed of a mixture of polypeptides ranging from approximately 40 to 330 kDa, mimicking those visualized by Western blotting of platelet lysates and releasates with anti-FV antibodies. The purified, platelet-derived protein expressed significant cofactor activity such that thrombin activation led to only a 2-3-fold increase in cofactor activity yet expression of a specific activity identical to that of purified, plasma-derived FVa. Physical and functional differences between the two cofactors were identified. Purified, platelet-derived FVa was 2-3-fold more resistant to activated protein C-catalyzed inactivation than purified plasma-derived FVa on the thrombin-activated platelet surface. The heavy chain subunit of purified, platelet-derived FVa contained only a fraction ( approximately 10-15%) of the intrinsic phosphoserine present in the plasma-derived FVa heavy chain and was resistant to phosphorylation at Ser(692) catalyzed by either casein kinase II or thrombin-activated platelets. MALDI-TOF mass spectrometric analyses of tryptic digests of platelet-derived FV peptides detected an intact heavy chain uniquely modified on Thr(402) with an N-acetylglucosamine or N-acetylgalactosamine, whereas Ser(692) remained unmodified. N-terminal sequencing and MALDI-TOF analyses of platelet-derived FV/Va peptides identified the presence of a full-length heavy chain subunit, as well as a light chain subunit formed by cleavage at Tyr(1543) rather than Arg(1545) accounting for the intrinsic levels of cofactor activity exhibited by native platelet-derived FVa. These collective data are the first to demonstrate physical differences between the two FV cofactor pools and support the hypothesis that, subsequent to its endocytosis by megakaryocytes, FV is modified to yield a platelet-derived cofactor distinct from its plasma counterpart.


Subject(s)
Factor V/chemistry , Platelet-Derived Growth Factor/chemistry , Blood Coagulation , Blood Platelets/metabolism , Factor V/isolation & purification , Factor V/metabolism , Humans , Platelet Activation , Platelet-Derived Growth Factor/isolation & purification , Platelet-Derived Growth Factor/metabolism , Structure-Activity Relationship , Thrombin/metabolism
9.
Blood ; 102(9): 3210-6, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-12816860

ABSTRACT

Severe factor V (FV) deficiency is a rare bleeding disorder, whose genetic bases have been characterized only in a limited number of cases. We investigated 6 unrelated patients with extremely reduced plasma FV levels, associated with a bleeding tendency ranging from moderately severe to severe. Clinical manifestations were substantially concordant with the previously established spectrum of hemorrhagic symptoms of the disease. Molecular analysis of FV gene identified 9 different mutations, 7 hitherto unknown, and 2 previously reported (Arg712ter and Tyr1702Cys). Four of 6 analyzed patients were compound heterozygotes, indicating the high allelic heterogeneity of this disease. Among novel mutations, 5 led to premature termination codons, because of nonsense (Arg1002ter, Arg1606ter, and Trp1854ter), or frameshift mutations (5127-5128insA and 6122-6123insAACAG). The remaining 2 were missense mutations (Cys472Gly and Val1813Met), located in FV A2 and A3 domains. Their effect on FV expression was studied by transient transfection experiments, demonstrating that the presence of each mutation impaired FV secretion. These data increase the number of severe FV deficiency-causing mutations by about 50%. The high number of "private" mutations identified in FV-deficient families indicates that full mutational screening of FV gene is still required for molecular diagnosis.


Subject(s)
Factor V Deficiency/genetics , Factor V/genetics , Mutation , Adolescent , Adult , Animals , COS Cells , Cloning, Molecular , Codon, Nonsense , Factor V/analysis , Factor V/isolation & purification , Family Health , Female , Frameshift Mutation , Hemorrhage/etiology , Humans , Male , Middle Aged , Mutation, Missense , Pedigree , Protein Structure, Tertiary , Transfection
10.
Biochemistry ; 38(35): 11448-54, 1999 Aug 31.
Article in English | MEDLINE | ID: mdl-10471296

ABSTRACT

Thrombin-activated factor Va exists as two isoforms, factor Va(1) and factor Va(2), which differ in the size of their light chains and their affinity for biological membranes. The heterogeneity of the light chain remained following incubation of factor Va with N-glycanase. However, we found that the factor V C2 domain, which contains a single potential glycosylation site at Asn-2181, was partially glycosylated when expressed in COS cells. To confirm the structural basis for factor Va(1) and factor Va(2), we mutated Asn-2181 to glutamine (N2181Q) and expressed this mutant using a B domain deletion construct (rHFV des B) in COS cells. Thrombin activation of N2181Q released a light chain with mobility identical to that of factor Va(2) on SDS-PAGE. The functional properties of purified N2181Q were similar to those of factor Va(2) in prothrombinase assays carried out in the presence of limiting concentrations of phosphatidylserine. The binding of human factor Va(1) and factor Va(2) to 75:25 POPC/POPS vesicles was also investigated in equilibrium binding assays using proteins containing a fluorescein-labeled heavy chain. The affinity of human factor Va(2) binding to POPC/POPS vesicles was approximately 3-fold higher than that of factor Va(1). These results indicate that partial glycosylation of factor V at asparagine-2181 is the structural basis of the light chain doublet and that the presence of this oligosaccharide reduces the affinity of factor Va for biological membranes.


Subject(s)
Asparagine/metabolism , Factor V/metabolism , Peptide Fragments/metabolism , Thromboplastin/metabolism , Animals , Asparagine/genetics , COS Cells , Cell Membrane/chemistry , Cell Membrane/metabolism , Factor V/biosynthesis , Factor V/genetics , Factor V/isolation & purification , Factor Va/chemistry , Factor Va/genetics , Factor Va/isolation & purification , Factor Va/metabolism , Glutamine/genetics , Glycoside Hydrolases/metabolism , Glycosylation , Humans , Mutagenesis, Site-Directed , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Protein Binding/genetics , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transfection
11.
Am J Hematol ; 61(4): 265-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10440914

ABSTRACT

The pathogenesis of venous thrombosis involves the interaction of genetic and environmental factors. In order to estimate the frequency of the factor V Leiden, the prothrombin G20210A, and the MTHFR C677T mutations in the Greek population, we analyzed 160 healthy Greek blood donors by PCR amplification and detected allele frequencies of 2.5%, 2.2%, and 35.3%, respectively. The allele frequencies were compared with reported frequencies of other populations of southern Europe. The identification of these common genetic risk factors for thrombosis should enable easy DNA diagnosis and carrier detection in a high proportion of cases and will contribute to a better understanding of the interaction of genetic and environmental risk factors.


Subject(s)
Blood Donors , Factor V/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Point Mutation , Prothrombin/genetics , Venous Thrombosis/genetics , Adult , Alleles , Factor V/isolation & purification , Female , Gene Frequency , Genetic Carrier Screening , Greece , Humans , Male , Methylenetetrahydrofolate Reductase (NADPH2) , Middle Aged , Oxidoreductases Acting on CH-NH Group Donors/isolation & purification , Polymerase Chain Reaction , Prevalence , Prothrombin/isolation & purification , Risk Factors , Venous Thrombosis/etiology
12.
Biochemistry ; 38(6): 1829-37, 1999 Feb 09.
Article in English | MEDLINE | ID: mdl-10026263

ABSTRACT

Factor VIIa, in complex with tissue factor (TF), is the serine protease responsible for initiating the clotting cascade. This enzyme complex (TF/VIIa) has extremely restricted substrate specificity, recognizing only three previously known macromolecular substrates (serine protease zymogens, factors VII, IX, and X). In this study, we found that TF/VIIa was able to cleave multiple peptide bonds in the coagulation cofactor, factor V. SDS-PAGE analysis and sequencing indicated the factor V was cleaved at Arg679, Arg709, Arg1018, and Arg1192, resulting in a molecule with a truncated heavy chain and an extended light chain. This product (FVTF/VIIa) had essentially unchanged activity in clotting assays when compared to the starting material. TF reconstituted into phosphatidylcholine vesicles was ineffective as a cofactor for the factor VIIa cleavage of factor V. However, incorporation of phosphatidylethanolamine in the vesicles had little effect over the presence of 20% phosphatidylserine. FVTF/VIIa was as sensitive to inactivation by activated protein C (APC) as thrombin activated factor V as measured in clotting assays or by the appearance of the expected heavy chain cleavage products. The FVTF/VIIa could be further cleaved by thrombin to release the normal light chain, albeit at a significantly slower rate than native factor V, to yield a fully functional product. These studies thus reveal an additional substrate for the TF/VIIa complex. They also indicate a new potential regulatory pathway of the coagulation cascade, i.e., the production of a form of factor V that can be destroyed by APC without the requirement for full activation of the cofactor precursor.


Subject(s)
Factor VIIa/metabolism , Factor V/metabolism , Factor Va/metabolism , Protein C/metabolism , Thromboplastin/metabolism , Animals , Cattle , Factor V/antagonists & inhibitors , Factor V/isolation & purification , Factor Va/isolation & purification , Humans , Hydrolysis , Macromolecular Substances , Protein C/physiology , Rabbits , Thrombin/pharmacology
13.
Thromb Haemost ; 78(1): 427-33, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9198191

ABSTRACT

Blood coagulation factor V plays an important role in the regulation of thrombin formation. Activation of factor V by traces of activated coagulation factors (thrombin, factor Xa or meizothrombin) yields factor Va, the non-enzymatic cofactor of the prothrombinase complex. Since factor Va accelerates prothrombin activation under physiological conditions more than 10(4)-fold it is not surprising that down-regulation of factor Va cofactor activity by the protein C pathway is a very effective way for maintaining the hemostatic balance. In this paper we have reviewed the present status of structural knowledge of factor V and Va, the molecular changes in factor V that occur during factor V activation, the function of factor Va in prothrombin activation and the molecular mechanism of inactivation of factor Va by APC. Although considerable insight in the structure-function relationship of factor V and Va has been achieved, the study of mutated factor V molecules obtained by recombinant DNA technology will undoubtedly resolve remaining questions. The latter is illustrated by the fact that the discovery of factor VaLeiden has significantly contributed to our present knowledge on the regulation of the cofactor activity of factor Va via the protein C pathway. It appears that modulation of the activity of APC by protein S and factor Xa will strongly affect the in vivo activity of this pathway. Factor V not only plays an important role in the regulation of the activity of the prothrombinase complex but also acts as cofactor in APC-mediated inactivation of factor VIIIa. This gives rise to a rather intricate mechanism of regulation of thrombin formation by APC that thus far has been mainly studied in model systems containing purified proteins. Thus, extensive studies in plasma will be required in order to get more insight in the in vivo regulation of thrombin formation via the protein C pathway.


Subject(s)
Factor V/chemistry , Factor Va/chemistry , Protein C/physiology , Prothrombin/metabolism , Arginine/chemistry , Factor V/isolation & purification , Factor V/physiology , Factor Va/physiology , Glutamine/chemistry , Humans , Mutation
14.
Biochemistry ; 36(11): 3319-30, 1997 Mar 18.
Article in English | MEDLINE | ID: mdl-9116010

ABSTRACT

The prothrombinase complex assembles through reversible interactions between factor Xa, factor Va and acidic phospholipid-containing membranes in the presence of calcium ions. This complex catalyses the conversion of prothrombin to thrombin through two proteolytic steps. We have used prethrombin 2 as a substrate analog for the first cleavage reaction of prothrombin activation (cleavage at Arg323-Ile324) catalyzed by the prothrombinase complex and have also relied on the known ability of prethrombin 2 to interact tightly but reversibly with fragment 2 or fragment 1.2. The kinetics of cleavage at Arg323-Ile324 have been assessed with these substrate analogs to investigate the contribution of cofactor-substrate interactions mediated by the fragment 2 domain to the ability of factor Va to enhance the catalytic efficiency of factor Xa within the prothrombinase complex. Initial velocity measurements indicated that the rate of prethrombin 2 cleavage by the factor Xa-PCPS binary complex was increased by a factor of approximately 1300 upon the addition of saturating concentrations of factor Va to assemble prothrombinase. Although the measured initial velocity was higher when either fragment 2 or fragment 1.2 was present, the factor Va-dependent enhancement in initial rate (2600- and 1500-fold) was comparable in each case. Steady state kinetic constants were obtained using prethrombin 2, prethrombin 2 plus fragment 2, and prethrombin 2 plus fragment 1.2 as substrates. For each substrate, the addition of saturating concentrations of factor Va to the Xa-PCPS binary complex led to increases in catalytic efficiency of between 1000 and 9000-fold. The kcat/Km for prethrombin 2 cleavage by prothrombinase was essentially identical to that obtained for prethrombin 2 saturated with fragment 2. Thus, comparable accelerating effects of factor Va are observed independent of the presence of the fragment 2 domain in the substrate. The results indicate that interactions between factor Va and the substrate mediated by the fragment 2 domain do not contribute in a dominant way to the ability of factor Va to enhance the catalytic efficiency of factor Xa within the prothrombinase complex.


Subject(s)
Factor V/metabolism , Factor Va/metabolism , Factor X/metabolism , Peptide Fragments/metabolism , Prothrombin/metabolism , Thrombin/metabolism , Animals , Arginine , Cattle , Factor V/isolation & purification , Factor Va/isolation & purification , Factor X/isolation & purification , Factor Xa/isolation & purification , Factor Xa/metabolism , Isoleucine , Kinetics , Liposomes , Peptide Fragments/isolation & purification , Phosphatidylcholines , Phosphatidylserines , Prothrombin/isolation & purification , Substrate Specificity
15.
Biochem J ; 313 ( Pt 2): 467-72, 1996 Jan 15.
Article in English | MEDLINE | ID: mdl-8573079

ABSTRACT

Resistance to activated protein C (APC), which is the most prevalent pathogenetic risk factor of thrombosis, is linked to a single point-mutation in the factor V (FV) gene, which predicts replacement of Arg (R) at position 506 with a Gln (Q). This mutation modifies one of three APC-cleavage sites in the heavy chain of activated FV (FVa), suggesting that mutated FVa (FVa:Q506) is at least partially resistant to APC-mediated degradation. To elucidate the molecular mechanisms of APC-resistance and to investigate the functional properties of FV in APC resistance, FV:Q506 was purified from an individual with homozygosity for the Arg to Gln mutation. Intact and activated FV:Q506 were demonstrated to convey APC resistance to FV-deficient plasma. Thrombin- or factor Xa-activated FV:Q506 were found to be approx. 10-fold less sensitive to APC-mediated degradation than normal FVa, at both high and low phospholipid concentrations. The degradation pattern observed on Western blotting suggested that FVa:Q506 was not cleaved at position 506. However, it was slowly cleaved at Arg306, which explains the partial APC sensitivity of FVa:Q506. FV is initially activated during clotting and then rapidly inactivated in a process which depends on the integrity of the protein C anticoagulant system. During clotting of APC-resistant plasma, FV:Q506 was activated in a normal fashion, but then only partially inactivated. In conclusion, the reduced sensitivity of FVa:Q506 to APC-mediated degradation is the molecular basis for the life-long hypercoagulable state which constitutes a risk factor for thrombosis in APC-resistant individuals.


Subject(s)
Arginine/genetics , Factor V/genetics , Glycine/genetics , Homozygote , Protein C/metabolism , Blood Coagulation , Blotting, Western , Factor V/isolation & purification , Factor V/metabolism , Humans , Hydrolysis , Protein S/metabolism , Thrombin/metabolism
16.
Blood ; 86(7): 2616-23, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7670105

ABSTRACT

The isolation and characterization of baboon plasma factor V (FV) were performed for the development of an in vivo model for studying factor V/Va physiology in nonhuman primates. Baboon FV was purified by immunoaffinity chromatography with an antihuman FV monoclonal antibody and exhibits a specific activity of 1,940 U/mg. Baboon FV activation by thrombin proceeds through two proteolytic pathways similar to those observed with human and bovine FV. Limited amino acid sequencing of FV and its thrombin activation fragments shows 95% identity with human and 79% identity with bovine FV. 125I-Factor V and a mixture of thrombin cleaved 125I-FV activation products were infused into normal male baboons and evaluated by blood sample radioactivity measurements and by autoradiography of plasma samples following resolution by gel electrophoresis. Factor V disappeared with a half-life (t1/2) of 12.98 +/- 1.85 hours and was cleared without obvious degradation of the molecule during circulation. The radioactivity associated with the thrombin activated FV mixture, which consisted of the Mr = 220,000 activation intermediate, the Mr = 150,000 activation peptide, the heavy chain (HC) and the light chain (LC) of FVa, was cleared in a nonlinear manner. The HC and LC were removed with t1/2 < 20 minutes. The apparent molecular weight (Mr) = 220,000 and Mr = 150,000 fragments were cleared with t1/2 > 6 hours and t1/2 > 30 hours, respectively.


Subject(s)
Factor V/metabolism , Models, Biological , Animals , Antibodies, Monoclonal , Autoradiography , Chromatography, Affinity/statistics & numerical data , Electrophoresis, Polyacrylamide Gel , Factor V/chemistry , Factor V/isolation & purification , Factor Va/metabolism , Humans , Iodine Radioisotopes , Isotope Labeling , Kinetics , Male , Metabolic Clearance Rate , Papio/blood , Protein C/metabolism , Sensitivity and Specificity , Sequence Analysis
18.
Blood ; 85(12): 3405-11, 1995 Jun 15.
Article in English | MEDLINE | ID: mdl-7780127

ABSTRACT

Gln506-factor V (FV) was purified from plasma of an individual homozygous for an Arg506Gln mutation in FV that is associated with activated protein C (APC) resistance. Purified Gln506-FV, as well as Gln506-FVa generated by either thrombin or FXa, conveyed APC resistance to FV-deficient plasma in coagulation assays. Clotting assay studies also suggested that APC resistance does not involve any abnormality in FV-APC-cofactor activity. In purified reaction mixtures, Gln506-FVa in comparison to normal FVa showed reduced susceptibility to APC, because it was inactivated approximately 10-fold slower than normal Arg506-FVa. It was previously reported that inactivation of normal FVa by APC involves an initial cleavage at Arg506 followed by phospholipid-dependent cleavage at Arg306. Immunoblot and amino acid sequence analyses showed that the 102-kD heavy chain of Gln506-FVa was cleaved at Arg306 during inactivation by APC in a phospholipid-dependent reaction. This reduced but measurable susceptibility of Gln506-FVa to APC inactivation may help explain why APC resistance is a mild risk factor for thrombosis because APC can inactivate both normal FVa and variant Gln506-FVa. In summary, this study shows that purified Gln506-FV can account for APC resistance of plasma because Gln506-FVa, whether generated by thrombin or FXa, is relatively resistant to APC.


Subject(s)
Factor V/genetics , Protein C/metabolism , Adult , Factor V/isolation & purification , Factor V/metabolism , Family , Glycine/genetics , Homozygote , Humans , Male , Point Mutation
19.
Blood ; 84(4): 1132-42, 1994 Aug 15.
Article in English | MEDLINE | ID: mdl-8049429

ABSTRACT

The procoagulant subcellular matrix of stimulated endothelial cells that contains tissue factor (TF) was used to investigate the mechanism by which TF pathway inhibitor (TFPI) inhibits thrombin formation initiated by TF/factor VIIa (FVIIa) under flow conditions. Purified coagulation factors VII, X, and V and prothrombin were perfused at a wall shear rate of 100 s-1 through a flow chamber containing a coverslip covered with matrix of cultured human umbilical vein endothelial cells. This resulted in a TF- and FVII-dependent FXa and thrombin generation as measured in the effluent at the outlet of the system. Inhibition of this TF/FVIIa-triggered thrombin formation by TFPI purified from plasma was dependent on the amount of TF present on the endothelial cell matrix. The rate of prothrombinase assembly and steady-state levels of thrombin formation were decreased by TFPI. Because persistent albeit decreased steady-state levels of thrombin formation occurred in the presence of TFPI, we conclude that plasma-TFPI does not inhibit FXa present in the prothrombinase complex. The addition of FIX and FVIII to perfusates containing FVII and FX increased the FXa generation on endothelial matrices, and counteracted the inhibition of thrombin formation on endothelial cell matrices by TFPI. Our data provide further evidence for the hypothesis that the rapid inactivation of TF/FVIIa by TFPI in combination with the absence of either FVIII or FIX causes the bleeding tendency of patients with hemophilia A or B.


Subject(s)
Endothelium, Vascular/physiology , Factor Xa/metabolism , Lipoproteins/pharmacology , Thrombin/metabolism , Thromboplastin/metabolism , Animals , Antibodies, Monoclonal , Cells, Cultured , Extracellular Matrix/physiology , Factor IX/isolation & purification , Factor IX/metabolism , Factor V/isolation & purification , Factor V/metabolism , Factor VII/isolation & purification , Factor VII/metabolism , Factor VIII/metabolism , Factor X/isolation & purification , Factor X/metabolism , Factor Xa Inhibitors , Humans , Kinetics , Lipoproteins/isolation & purification , Mice/immunology , Models, Biological , Prothrombin/isolation & purification , Prothrombin/metabolism , Recombinant Proteins/metabolism , Time Factors , Umbilical Veins
20.
Thromb Haemost ; 70(6): 963-9, 1993 Dec 20.
Article in English | MEDLINE | ID: mdl-8165619

ABSTRACT

Monocyte-derived macrophages, focal to initiation and progression of atherosclerosis, have been implicated in thrombotic complication of this disease. In the present study we demonstrated tissue factor based procoagulant activity in cultured macrophages from the White Carneau pigeon following endotoxin (1-2 micrograms/ml) stimulation. This macrophage procoagulant activity paralleled activity obtained with pigeon brain homogenate. We used Enzyme-Linked Coagulation Assay (ELCA), an ultrasensitive microtiter plate assay, to measure procoagulant activity in these cells. Through the use of clotting factors purified from pigeon plasma, procoagulant activity could be detected with as few as 1-3 cells. Tissue factor antigen, detected through the use of immunogold labelling in conjunction with a polyclonal antibody which was highly specific to human tissue factor, was distributed uniformly over the plasma membrane of the endotoxin-stimulated cells. These studies suggest that this procoagulant activity might play an important role in the pathobiology of atherosclerosis in White Carneau pigeons by initiating fibrin polymerization and thus leading to thrombotic complications of the disease.


Subject(s)
Columbidae/blood , Macrophages/chemistry , Monocytes/cytology , Thromboplastin/analysis , Animals , Blood Coagulation Tests , Cells, Cultured , Endotoxins/pharmacology , Factor V/isolation & purification , Factor VII/isolation & purification , Factor X/isolation & purification , Humans , Immunohistochemistry , Monocytes/drug effects , Prothrombin/isolation & purification , Sensitivity and Specificity , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...