Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Appl Microbiol Biotechnol ; 97(14): 6263-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23129183

ABSTRACT

Laccases produced by white rot fungi have been extensively evaluated for their potential to decolorize textile wastewaters which contain salts like sodium chloride and sodium sulfate. The effect of sodium chloride and sodium sulfate on Trametes versicolor laccase during the decolorization of an anthraquinone dye (Reactive Blue 19) and the oxidation of 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) were evaluated by steady-state kinetic analysis. The results showed that, while sodium sulfate did not affect laccase activity, sodium chloride inhibited both ABTS oxidation and dye decolorization. However, the type of inhibition was substrate-dependent: it was hyperbolic, noncompetitive with ABTS and parabolic, noncompetitive with Reactive Blue 19. Furthermore, the results suggested that two chlorides may bind to laccase in the presence of the dye unlike recent inhibition models which suggest that there is only one inhibition site. This investigation is the first to provide evidence for and to propose a two-site model of laccase inhibition, providing new insight into NaCl inhibition of laccase. The proposed model is also useful to predict decolorization rates in the presence of sodium chloride and to determine operating conditions that will minimize inhibition.


Subject(s)
Anthraquinones/metabolism , Benzothiazoles/chemistry , Coloring Agents/metabolism , Enzyme Inhibitors/chemistry , Fungal Proteins/metabolism , Laccase/metabolism , Sodium Chloride/chemistry , Sulfonic Acids/chemistry , Trametes/enzymology , Anthraquinones/chemistry , Biodegradation, Environmental , Coloring Agents/chemistry , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/chemistry , Kinetics , Laccase/antagonists & inhibitors , Laccase/chemistry , Oxidation-Reduction
2.
Haemophilia ; 18(3): e316-22, 2012 May.
Article in English | MEDLINE | ID: mdl-21933309

ABSTRACT

Individuals with haemophilia A exhibit bleeding tendencies that are not always predicted by their factor (F)VIII level. It has been suggested that bleeding in haemophilia is due not only to defective prothrombin activation but also aberrant fibrinolysis. Thrombin activatable fibrinolysis inhibitor (TAFI) activation was measured in tissue factor (TF)-initiated blood coagulation in blood samples of 28 haemophiliacs and five controls. Reactions were quenched over time with FPRck and citrate and assayed for TAFIa and thrombin-antithrombin (TAT). The TAFIa potential (TP), TAFI activation rate and the TAFIa level at 20 min (TAFIa(20 min)) was extracted from the TAFI activation progress curve. In general, the time course of TAFI activation follows thrombin generation regardless of FVIII activity and as expected the rate of TAFI activation and TP decreases as FVIII decreases. The magnitude of TP was similar among the control subjects and subjects with <11% FVIII. In severe subjects with <1% FVIII at the time of blood collection, the TAFIa(20 min) was inversely and significantly correlated with haemarthrosis (-0.77, P = 0.03) and total bleeds (-0.75, P = 0.03). In all cases, TAFIa(20 min) was more strongly correlated with bleeding than TAT levels at 20 min. Overall, this study shows that TAFI activation in whole blood can be quantified and related to the clinical bleeding phenotype. Measuring TAFIa along with thrombin generation can potentially be useful to evaluate the differential bleeding phenotype in haemophilia A.


Subject(s)
Carboxypeptidase B2/metabolism , Fibrinolysis/physiology , Hemophilia A/enzymology , Blood Coagulation/physiology , Enzyme Activation/physiology , Hemarthrosis/enzymology , Hemarthrosis/physiopathology , Hemorrhage/enzymology , Hemorrhage/physiopathology , Humans , Phenotype , Thrombin/metabolism
3.
J Clin Invest ; 74(6): 2219-25, 1984 Dec.
Article in English | MEDLINE | ID: mdl-6439744

ABSTRACT

An experimental animal model of disseminated intravascular coagulation (DIC) induced by the co-infusion of coagulant-active phospholipid and activated Factor X (Factor Xa) is described. The infusion of Factor Xa at a dose of 6.6 X 10(-12) mol/kg with phosphatidylcholine/phosphatidylserine (PCPS) lipid vesicles at a dose of 4.0 X 10(-8) mol/kg was associated with significant falls in the levels of fibrinogen and Factors V and VIII, and a bleeding diathesis developed. Assays of Factors V and VIII were performed by a one-stage prothrombin time and activated partial thrombin time system, respectively. In additional experiments, the effect of the same dose combination of Factor Xa/PCPS on Factor V kinetics was studied by preinfusing 125I-labeled Factor V. After Factor Xa/PCPS infusion, Factors VIII and V were reduced at 2 min by 90 and 50% of the preinfusion levels, respectively, and at 1 h by 80 and 75%, respectively. During the same period, there was little change in the total circulating radioactivity. Autoradiography indicated small but detectable levels of circulating proteolytic products of Factor V that comigrated with peptides obtained by the incubation of Factor V with Factor Xa and activated protein C. The majority of radioactivity remained associated with the intact single-chain precursor Factor V. These observations suggested maintenance of the precursor pool after the onset of DIC. This was confirmed by performing two-stage assays of Factors V and VIII, whereby each was completely converted to the active cofactor, i.e., Va and VIII:Ca, by preincubation of the test sample with thrombin before assaying in a one-stage system as before. The Factor V levels assayed by the two-stage procedure did not change appreciably over 1 h. The Factor VIII levels fell but corrected within 1 h at a time when the level measured by a one-stage assay remained depressed. These results indicate that in the dog, infusion of Factor Xa/PCPS induces changes characteristic of DIC, and this is associated with the appearance of Factor V peptides characteristic of the expression of Factor Xa and activated protein C-like activities. The differences noted between the one-stage and two-stage assays suggest that the one-stage assay is measuring the activated fraction of each cofactor and not the total level of the available precursor for each activated species. The results suggest a close correlation between the activated fraction of both cofactors and the hemostatic abnormality that occurs in DIC.


Subject(s)
Disease Models, Animal , Disseminated Intravascular Coagulation/metabolism , Factor VIII/metabolism , Factor V/metabolism , Animals , Disseminated Intravascular Coagulation/chemically induced , Dogs , Factor X , Factor Xa , Fibrinogen/metabolism , Fibrinolysis , Glycoproteins/metabolism , Heparin/pharmacology , Kinetics , Liposomes , Phosphatidylcholines , Phosphatidylserines , Protein C
4.
J Clin Invest ; 65(3): 666-74, 1980 Mar.
Article in English | MEDLINE | ID: mdl-6444419

ABSTRACT

A woman, aged 68, with multiple myeloma (immunoglobulin[Ig]A kappa type) developed an anticoagulant with properties suggestive of heparin. The anticoagulant prolonged the thrombin time but not the reptilase time and was resistant to boiling, proteolytic enzyme digestion, and trichloracetic acid precipitation. The thrombin time was corrected by the addition (in vitro) of protamine sulfate or the addition of purified platelet Factor 4 (PF4) to the plasma. The anticoagulant was isolated by PF4-Sepharose affinity chromatography and analyzed in terms of its molecular weight, uronic acid, and amino acid composition. The proteoglycan isolated had a mol wt of 116,000 and appears to consist of two 38,000 dalton polysaccharide units interconnected by peptide material totaling 39,000 daltons. Electrophoretic analysis of the pronase digested peptidoglycan using the lithium acetate-agarose technique suggested the material was of the heparan sulfate type. The peptidoglycan had about one-tenth the specific activity of commercially available heparin on a weight basis. The isolated proteoglycan was indistinguishable from commercial heparin when analyzed in terms of its ability to act as a cofactor in the antithrombin III inhibition of thrombin.


Subject(s)
Blood Coagulation , Glycosaminoglycans/blood , Heparitin Sulfate/blood , Multiple Myeloma/blood , Proteoglycans/blood , Aged , Amino Acids/analysis , Female , Heparitin Sulfate/isolation & purification , Humans , Molecular Weight , Proteoglycans/isolation & purification
5.
J Clin Invest ; 66(5): 934-40, 1980 Nov.
Article in English | MEDLINE | ID: mdl-6776145

ABSTRACT

A dysprothrombin designated prothrombin Quick, is isolated from the plasma of an individual with < 2% of normal functional prothrombin activity and 34% of the normal prothrombin level by immunologic assay. With Factor Xa or taipan snake venom as activators, a fragmentation pattern identical to that of normal prothrombin is observed on gel electrophoresis in dodecylsulfate. This evidence combined with the observed barium citrate adsorption of prothrombin Quick and the low activity suggests that the defect in prothrombin Quick is in the thrombin portion of the molecule. Thrombin Quick is isolated and comigrates with thrombin on dodecyl sulfate gel electrophoresis, either reduced or nonreduced. The activity of thrombin Quick on several biological substrates of thrombin is investigated. Relative to normal thrombin, thrombin Quick is 1/200 as active on fibrinogen and 1/20-1/50 as effective in activating Factors V and VIII and aggregating platelets. A complex with antithrombin III is detected by dodecyl sulfate gel electrophoresis. Further investigation with the active site titrant, dansylanginine-N-(3-ethyl-1,5-pentanediyl)amide showed that the thrombin Quick preparation has the same affinity for the titrant as thrombin, but apparently only 40% active sites per mole protein are titrable.


Subject(s)
Hypoprothrombinemias/blood , Thrombin/genetics , Antithrombin III/metabolism , Arginine/analogs & derivatives , Arginine/metabolism , Calcium/pharmacology , Chromatography, Gel , Dansyl Compounds/metabolism , Electrophoresis, Polyacrylamide Gel , Factor VIII/metabolism , Factor X/pharmacology , Factor Xa , Fibrinogen/metabolism , Humans , Phospholipids/pharmacology , Thrombin/metabolism , Thrombin/physiology
6.
J Clin Invest ; 99(10): 2323-7, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9153272

ABSTRACT

Recently, it has been shown that Factor XI can be activated by thrombin, and that Factor XIa significantly contributes to the generation of thrombin via the intrinsic pathway after the clot has been formed. This additional thrombin, generated inside the clot, was found to protect the clot from fibrinolysis. A plausible mechanism for this inhibitory effect of thrombin involves TAFI (thrombin-activatable fibrinolysis inhibitor, procarboxypeptidase B) which, upon activation, may inhibit fibrinolysis by removing carboxy-terminal lysines from fibrin. We studied the role of Factor XI and TAFI in fibrinolysis using a clot lysis assay. The lysis time was decreased twofold when TAFI was absent, when TAFI activation was inhibited by anti-TAFI antibodies, or when activated TAFI was inhibited by the competitive inhibitor (2-guanidinoethylmercapto)succinic acid. Inhibition of either TAFI activation or Factor XIa exhibited equivalent profibrinolytic effects. In the absence of TAFI, no additional effect of anti-Factor XI was observed on the rate of clot lysis. We conclude that the mechanism of Factor XI-dependent inhibition of fibrinolysis is through the generation of thrombin via the intrinsic pathway, and is dependent upon TAFI. This pathway may play a role in determining the fate of in vivo formed clots.


Subject(s)
Carboxypeptidases/metabolism , Factor XI/metabolism , Factor XIa/metabolism , Fibrinolysis , Thrombin/physiology , Antibodies, Monoclonal , Carboxypeptidase B2 , Carboxypeptidases/isolation & purification , Carboxypeptidases/pharmacology , Chromatography, Affinity , Fibrinolysis/drug effects , Humans , Kinetics , Succinates/pharmacology , Tissue Plasminogen Activator/metabolism
7.
J Clin Invest ; 77(2): 405-15, 1986 Feb.
Article in English | MEDLINE | ID: mdl-3944265

ABSTRACT

A coagulation Factor V inhibitor developed in a man 75 yr of age in association with an anaplastic malignancy and drug treatment (including the aminoglycoside antibiotic, gentamicin). The patient did not bleed abnormally, despite both surgical challenge and plasma Factor V activity of less than 1%. The inhibited plasma had grossly prolonged prothrombin and activated partial thromboplastin times, but a normal thrombin time. Mixing studies indicated progressive coagulation inhibition with normal plasma, but not with Factor V-deficient plasma, and reversal of coagulation inhibition by the addition of bovine Factor V to the patient's plasma. 1 ml of patient plasma inhibited the Factor V activity of 90 ml of normal human plasma. The inhibitor was isolated by sequential affinity chromatography on protein A-Sepharose and Factor V-Sepharose. The IgG isolate markedly inhibits the activity of prothrombinase assembled from purified Factors Xa and Va, calcium ion, and phospholipid vesicles, and partially inhibits prothrombinase assembled from purified Factor Xa, calcium ion, and normal platelets. The Factor V of platelets, however, appears relatively inaccessible to the antibody, inasmuch as platelets isolated from whole blood supplemented for 8 h with the antibody functioned normally with respect to platelet Factor V-mediated prothrombinase function. The absence of obvious hemorrhagic difficulties in the patient, the total inhibition of plasma Factor V by the inhibitor, and the apparent inaccessibility of platelet Factor V to the inhibitor specifically implicate platelet Factor V in the maintenance of hemostasis.


Subject(s)
Autoantibodies/isolation & purification , Factor V/antagonists & inhibitors , Factor Xa , Aged , Autoantibodies/physiology , Blood Coagulation , Chromatography, Affinity , Electrophoresis, Polyacrylamide Gel , Factor V/immunology , Factor V/metabolism , Factor Va , Factor X/metabolism , Humans , Immunoglobulin G/physiology , Male , Partial Thromboplastin Time , Prothrombin Time
8.
J Thromb Haemost ; 5(6): 1250-6, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17266698

ABSTRACT

BACKGROUND: Elevated plasma fibrinogen is a well known risk factor for cardiovascular disease. The mechanistic rationale for this is not known. OBJECTIVES: These studies were carried out to determine the fibrinogen concentration dependencies of clotting and lysis times and thereby determine whether these times rationalize the correlation between an increased risk of cardiovascular disease and elevated plasma fibrinogen. METHODS: The time courses of clot formation and lysis were measured by turbidity in systems comprising a) fibrinogen, thrombin and plasmin, or b) fibrinogen, thrombin, plasminogen and t-PA, or c) plasma, thrombin and t-PA. From the lysis times, k(cat) and K(m) values for plasmin action on fibrin were determined. RESULTS: The time to clot increased linearly from 2.9 to 5.6 minutes as the fibrinogen concentration increased from 1 to 9 microM and did not increase further as the fibrinogen concentration was raised to 20 microM. In contrast, the clot lysis time increased linearly over the input fibrinogen concentration range of 2 to 20 microM. A similar linear trend was found in the two systems with t-PA and plasminogen. Apparent K(m) and k(cat) values for plasmin were 1.1 +/- 0.6 microM and 28 +/- 2 min(-1), respectively. K(m) values for plasmin in experiments initiated with t-PA and plasminogen were 1.6 +/- 0.2 microM in the purified system and 2.1 +/- 0.9 microM in plasma. CONCLUSION: As the concentration of fibrinogen increases, especially above physiologic level, the balance between fibrinolysis and clotting shifts toward the latter, providing a rationale for the increased risk of cardiovascular disease associated with elevated fibrinogen.


Subject(s)
Blood Coagulation/physiology , Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Fibrinogen/metabolism , Hemolysis/physiology , Blood Coagulation/drug effects , Fibrin/metabolism , Fibrinogen/chemistry , Hemolysis/drug effects , Humans , In Vitro Techniques , Kinetics , Models, Cardiovascular , Nephelometry and Turbidimetry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Plasminogen/metabolism , Plasminogen/pharmacology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Tissue Plasminogen Activator/metabolism , Tissue Plasminogen Activator/pharmacology , alpha-2-Antiplasmin/metabolism , alpha-2-Antiplasmin/pharmacology
9.
J Thromb Haemost ; 1(6): 1258-63, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12871328

ABSTRACT

Antithrombin and its cofactor, heparin, target both the product of prothrombin activation by prothrombinase, thrombin, as well as the enzyme responsible for the reaction, factor (F)Xa. These studies were carried out to quantify the effects of each of the prothrombinase components on the half-life of FXa in the presence of antithrombin and the low-molecular-weight heparins (enoxaparin, Aventis, Laval, Quebec, Canada) or the heparin pentasaccharide (fondaparinux, Organon Sanofi-Synthelabo, Cypress, TX, USA). Experiments were carried out using a recombinant form of prothrombin in which the active site serine has been mutated to cysteine and subsequently labeled with fluorescein. This mutant allowed calculation of the second order rate constant for inhibition of FXa by antithrombin in such a way that competition for antithrombin by thrombin is eliminated and competition for FXa by prothrombin is accounted for. Intrinsic rate constants for the inhibition of FXa by antithrombin-enoxaparin and antithrombin-fondaparinux, in the presence of the various prothrombinase components, were calculated. Addition of phospholipid had no significant effect on the second order rate constant for inhibition of FXa by antithrombin, while addition of FVa appeared to be mildly protective. Further addition of prothrombin however, caused profound protection of FXa, increasing its half-life from 1.1 to 353 s in the case of fondaparinux, and from 0.4 to 42 s in the case of enoxaparin. Similar results were reported for unfractionated heparin previously [1]. Therefore, in the presence of unfractionated heparin, fondaparinux, or enoxaparin, prothrombinase is profoundly protected from antithrombin.


Subject(s)
Antithrombin III/pharmacology , Enoxaparin/pharmacology , Factor V/drug effects , Factor Xa/drug effects , Polysaccharides/pharmacology , Binding Sites/genetics , Catalysis , Drug Therapy, Combination , Factor Xa/metabolism , Fondaparinux , Half-Life , Humans , Kinetics , Models, Theoretical , Mutation , Prothrombin/genetics , Recombinant Proteins/genetics
10.
J Thromb Haemost ; 2(11): 1969-79, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15550029

ABSTRACT

Regulation of mRNA stability has emerged as a major control point in eukaryotic gene expression. The abundance of a particular mRNA can be rapidly regulated in response to a stimulus by altering the stability of existing translatable transcripts rather than by altering the rate of transcription initiation. Alternative polyadenylation of transcripts during mRNA processing can be important in determining transcript abundance if the different forms of mRNA possess different stabilities or translatability. The mRNA transcript encoding thrombin activable fibrinolysis inhibitor (TAFI) is an attractive candidate for regulation of mRNA stability because of the relatively long length of its 3'-untranslated region and because the transcript can be polyadenylated at three different sites. As well, we have previously reported that treatment of HepG2 cells with interleukins (IL) - 1beta and - 6 destabilizes the endogenous TAFI mRNA expressed in this cell line. In the current study, we report that the TAFI 3'-untranslated region contains cis-acting instability element(s) and that these elements in fact determine the intrinsic stability of the TAFI transcript. Moreover, we found that the three different polyadenylated mRNA forms have different intrinsic stabilities, with the mRNA half-life increasing from the longest to the shortest transcript. Interestingly, treatment with IL-1beta plus IL-6 not only resulted in a 2-fold decrease in stability of the transcript produced using the 3'-most polyadenylation site but also resulted in profound shifts in the relative abundances of the respective polyadenylated forms through changes in the frequency of utilization of the three polyadenylation sites. As such, in the presence of IL-1beta and IL-6, the longest transcript is over a thousand times more abundant than the two shorter transcripts whereas in the absence of the stimulus it comprises only 1% of the total TAFI transcripts.


Subject(s)
Carboxypeptidase B2/genetics , Gene Expression Regulation , RNA Stability/physiology , RNA, Messenger/chemistry , 3' Untranslated Regions/genetics , 3' Untranslated Regions/physiology , Cell Line, Tumor , Gene Expression Regulation/drug effects , Half-Life , Humans , Interleukin-1/pharmacology , Interleukin-6/pharmacology , Polyadenylation/drug effects , Polyadenylation/genetics , RNA Stability/drug effects , RNA, Messenger/drug effects
11.
J Thromb Haemost ; 2(9): 1535-44, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15333027

ABSTRACT

Summary. The biphasic waveform is an early marker of disseminated intravascular coagulation (DIC). Neutrophil elastase (NE) cleaves coagulation factors; thus, elevated elastase levels or its dysregulation by alpha-1-protease inhibitor (Alpha1PI) may be linked to DIC. Time courses over a period were determined for factors associated with NE and coagulation in 14 Intensive Care Unit patients with a biphasic waveform who developed DIC. The data were analyzed using a random coefficient linear regression model to predict the variables' mean values on day 0 and their mean rates of change over the period in which the biphasic waveform appeared. The biphasic waveform was normal on day 0, maximized on day 1, and approached normal again by day 4. Alpha1PI/NE complex levels were 2.5-fold greater than normal for the entire period. The A1PI activity, antigen, and specific activity levels were normal on day 0 and increased thereafter by 21.0, 10.5, and 8.9% of normal per day, respectively. Factor II, V, VII, IX, and X activity levels were, respectively, 57, 46, 46, 77, and 46% of normal on day 0, whereas factor VIII and fibrinogen levels were normal. All coagulation factor levels trended upward with time but not significantly. The prothrombin time, but not the activated partial thromboplastin time, was prolonged, and the platelet counts and hematocrits were below normal on day 0 and remained so thereafter. We conclude that events associated with neutrophil activation, elastase release, and perturbations of coagulation precede both the appearance of the biphasic waveform and the diagnosis of DIC in these patients.


Subject(s)
Blood Coagulation , Disseminated Intravascular Coagulation/blood , Disseminated Intravascular Coagulation/enzymology , Leukocyte Elastase/blood , Adult , Aged , Aged, 80 and over , Blood Coagulation Factors/metabolism , Critical Care , Disseminated Intravascular Coagulation/etiology , Female , Humans , Male , Middle Aged , Risk Factors , Time Factors , alpha 1-Antitrypsin/metabolism
12.
Methods Enzymol ; 222: 260-80, 1993.
Article in English | MEDLINE | ID: mdl-8412798

ABSTRACT

The approaches described in this article have resulted in an increased understanding of the reaction steps involved in the stabilization and assembly of the prothrombinase complex. Because prothrombinase is considered an archetype for some of the other coagulation complexes, the quantitative information derived from these studies (Table I) provides the framework for future studies of prothrombinase and suggests experimental approaches for studies of the other analogous coagulation reactions.


Subject(s)
Factor V/metabolism , Factor X/metabolism , Animals , Calorimetry , Cattle , Chromatography, Gel/methods , Factor V/biosynthesis , Factor V/isolation & purification , Factor Va/isolation & purification , Factor Va/metabolism , Factor X/biosynthesis , Factor X/isolation & purification , Factor Xa/isolation & purification , Factor Xa/metabolism , Fluorescence Polarization/methods , Fluorescent Dyes , Indicators and Reagents , Kinetics , Mathematics , Spectrometry, Fluorescence/methods
13.
Methods Enzymol ; 215: 329-60, 1992.
Article in English | MEDLINE | ID: mdl-1331708

ABSTRACT

The assembly and function of the prothrombinase complex on the bovine and human platelet membrane is mediated through binding interactions in which factor Va bound to the platelet surface forms at least part of the "receptor" for factor Xa in a 1:1 stoichiometric complex. A model depicting these binding interactions is shown in Fig. 12. Data from our laboratory indicate that the prothrombinase catalyst assembles in an analogous manner on the surface of monocytes, lymphocytes, neutrophils, and well-defined phospholipid vesicles employed in model systems. The 74,000-Da subunit of factor Va, component E, which mediates the binding of factor Va to either bovine platelets, human monocytes, or phospholipid vesicles, is shown binding to the cell membrane through its putative "receptor." The 94,000-Da subunit of factor Va, component D, is associated with the membrane surface through its metal ion-dependent interaction with component E. Factor Va forms at least part of the receptor that mediates the binding of factor Xa to an appropriate membrane surface, because component E has been shown to contribute significantly to the interaction of factor Xa with either the platelet, monocyte, or vesicle membrane surface. Our data do not preclude the possibility that component D contributes to the binding of factor Xa and the function of the prothrombinase complex. Component D appears to be important for several reasons. Cleavage of component D by activated protein C results in the complete loss of factor Va cofactor activity. An interaction between factor Xa and component D is implied from the observation that factor Xa protects factor Va from activated protein C inactivation. Furthermore, the binding of factor Xa to platelet-bound factor Va results in the time-dependent cleavage of components D and D'. Because component D is not required absolutely for prothrombinase complex assembly, we would speculate that it may be important in mediating prothrombin binding (depicted as a three-domain molecule) and increasing the catalytic efficiency of the enzymatic complex.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Blood Platelets/metabolism , Factor Va/metabolism , Factor Xa/metabolism , Platelet Membrane Glycoproteins , Receptors, Cell Surface/metabolism , Animals , Cattle , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Humans , Iodine Radioisotopes , Kinetics , Macromolecular Substances , Mathematics , Models, Structural , Prothrombin/metabolism , Radioisotope Dilution Technique , Receptors, Cell Surface/isolation & purification , Thromboplastin/metabolism
14.
Thromb Haemost ; 70(1): 80-6, 1993 Jul 01.
Article in English | MEDLINE | ID: mdl-8236120

ABSTRACT

The surface of the platelet expresses procoagulant phospholipids that bind coagulation factors and contribute to the procoagulant activity of the cell. The evidence, however, also suggests that platelets may possess specific receptors for coagulation Factors V(a) and VIII(a). Examination of a limited set of the available data could lead to the controversial position that the procoagulant surface of platelets is defined exclusively by either procoagulant phospholipids, or specific receptors for the cofactors and proteases. At the present time insufficient data exist to exclusively support one or the other of these possibilities. Further work, however, should indicate clearly whether the putative receptors exist and what role they play, either alone or in concert with surface phospholipids, in the expression and regulation of platelet procoagulant activity.


Subject(s)
Blood Platelets/chemistry , Platelet Membrane Glycoproteins/analysis , Receptors, Cell Surface/analysis , Biotransformation/physiology , Humans , Phospholipids/blood , Prothrombin/metabolism , Templates, Genetic
15.
Thromb Haemost ; 65(3): 280-5, 1991 Mar 04.
Article in English | MEDLINE | ID: mdl-1904654

ABSTRACT

The binding of t-PA to fibrin is mediated both by its "finger" (F) and its "kringle 2" (K2) domain. In addition, these domains are involved in the stimulation of t-PA activity by fibrin. We analyzed the kinetic characteristics of Glu-plasminogen activation by t-PA and a set of t-PA deletion mutants in the absence and the presence of desA-fibrin. In the absence of desA-fibrin, the activity of t-PA (variants) is determined by the presence of the protease domain, irrespective of the composition of the amino-terminal heavy chain. In the presence of the cofactor desA-fibrin, the activity of t-PA (variants) is dependent on the domain composition of the heavy chain. The activity of t-PA is stimulated 2,400 fold by desA-fibrin, whereas the activity of the mutant lacking the K1 domain (del. K1) increases 936 fold in the presence of this cofactor. Mutants lacking either the K2 domain (del. K2) or the F domain (del. F) exhibit an enhanced activity upon desA-fibrin addition of 200 and 210 fold, respectively. DesA-fibrin has no stimulatory effect on the activity of the mutant containing only the serine-protease domain (del.FE K1 K2) nor on the activity of the variant containing only the K1 domain and the serine-protease domain (del. FE K2). Furthermore, we determined the relative fibrin affinity of each t-PA variant, which is similarly dictated by the composition of the heavy chain.


Subject(s)
Plasminogen/drug effects , Tissue Plasminogen Activator/pharmacology , Amino Acid Sequence , Fibrin/metabolism , Kinetics , Molecular Sequence Data , Mutation , Protein Binding , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism
16.
Thromb Haemost ; 63(3): 476-81, 1990 Jun 28.
Article in English | MEDLINE | ID: mdl-2402750

ABSTRACT

Parameters of the fibrinolytic system were studied in a primate model where the generation of thrombin was promoted in vivo. The procoagulant stimulus used was a combination of human factor Xa in combination with phosphatidylcholine/phosphatidylserine lipid vesicles (PCPS) as the source of coagulant active phospholipid. The dosage of each component was formulated to provide a gradation of thrombin generating potential assessed prior to in vivo study in an in vitro clotting assay. These ranged from 25.25-36.60 pMole/kg (factor Xa) and 18.85-56.30 nMole/kg (PCPS). In each case, the ratio of the dose of factor Xa/PCPS was maintained at 0.65 (pMole factor Xa/nMole PCPS). Individual dosage combinations producing recalcification clotting times in vitro of 15, 20, 25 and 30 s were used in detailed in vivo studies. Previous studies in dogs had confirmed the thrombin generating potential of factor Xa/PCPS infusions and demonstrated an associated activation of protein C and increased fibrinolytic activity. This has now been extensively characterized in the chimpanzee as follows: 10 min after the infusion of the highest dose (36.6 pMole factor Xa/56.3 nMole PCPS kg bodyweight), the level of circulating t-PA had risen to 900 ng/ml (antigen), 885 IU/ml (functional). Dosage was observed with the lowest dose of 12.25 pMole factor Xa and 18.85 nMole PCPS being associated with relatively minor increases in circulating t-PA activity. There were no changes in u-PA at any dosage during the full time course of the experimental period (90 min). Plasminogen activation was also apparent with alpha-2 antiplasmin levels falling to 30-40% of pre-infusion levels at the highest dosages.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Fibrinolysis/physiology , Pan troglodytes/blood , Thrombin/biosynthesis , Animals , Blood Coagulation Tests , Factor Xa , Phosphatidylcholines , Phosphatidylserines , Plasminogen Inactivators/analysis , Reference Values
17.
Thromb Haemost ; 69(5): 448-53, 1993 May 03.
Article in English | MEDLINE | ID: mdl-7686692

ABSTRACT

Activation and inactivation of protein C during the clinical course of disseminated intravascular coagulation (DIC) was studied in three patients by qualitative (Western blotting) and quantitative (ELISA) analysis and the intensity of procoagulant activity monitored by the measurement of thrombin and factor Xa antithrombin III complexes. In one patient, inhibitor complexes of APC with protein C inhibitor (PCI) and alpha 1-antitrypsin (alpha 1-AT) were observed and the latter predominated at presentation. Both disappeared during the development of remission but the loss of alpha 1-AT complexes preceded PCI complexes which on Western blotting appeared to increase in intensity prior to disappearance. The two other patients bled to death from uncontrollable haemorrhage. In both cases, APC/inhibitor complexes with alpha 2-macroglobulin (alpha 2-M) in addition to PCI and alpha 1-AT were detected and persisted until death. Although PCI appeared to be the primary inhibitor in all three cases, alpha 1-antitrypsin and particularly alpha 2-macroglobulin appeared to assume greater roles in the two fatal cases. These data are similar to previous findings in an experimental animal model of DIC that suggested that alpha 2-macroglobulin and alpha 1-antitrypsin become more important inhibitors of APC as the primary inhibitor PCI is consumed in the face of a sustained procoagulant challenge.


Subject(s)
Disseminated Intravascular Coagulation/blood , Plasminogen Inactivators/metabolism , Protein C/antagonists & inhibitors , alpha 1-Antitrypsin/metabolism , alpha-Macroglobulins/metabolism , Adult , Antithrombin III/analysis , Blood Coagulation Tests , Cerebral Hemorrhage/blood , Enzyme Activation , Female , Humans , Male , Middle Aged , Peptide Hydrolases/analysis , Postoperative Complications/blood , Protein C/metabolism , Protein C Inhibitor , Puerperal Disorders/blood
18.
Thromb Res ; 47(3): 341-52, 1987 Aug 01.
Article in English | MEDLINE | ID: mdl-2442830

ABSTRACT

Anticoagulant properties of three sulfated compounds prepared from xylans isolated from corn cobs, larchwood and oatspelts were compared with heparin and sodium pentosan polysulfate (SP-54) by studying their effects on activated partial thromboplastin time (APTT), prothrombin time (PT) and thrombin time (TT) using pooled normal human plasma. These compounds were more effective than SP-54 in delaying coagulation by all the three procedures while oatspelts xylan sulfate was as effective as heparin in inhibiting APTT and PT and more effective than heparin in inhibiting TT on a molar basis. The sulfated xylans were more effective than heparin or SP-54 in potentiating the AT-III inhibition of amidolysis of H-D-Phe-Pip-Arg-pNa (S-2238) by thrombin (IIa) or amidolysis of Bz-Ile-Glu-Gly-Arg-pNa (S-2222) by Xa. Study of the high affinity binding of the xylan sulfates to AT-III-Sepharose column showed that the amount of the xylan sulfate recovered in the eluates from this peak was greatly increased with an increase in molecular weight (MW). A buffered mixture of IIa, AT-III and dansylarginine N-(3-ethyl-1,5-pentanediyl) amide (DAPA) was used to study the inactivation of IIa by AT-III. Larchwood xylan sulfate (2-10 micrograms) was found to accelerate this inactivation which was neutralized by human platelet factor 4 (PF4). The results also suggested an interaction between larchwood xylan sulfate and IIa which may potentiate an interaction between AT-III and IIa.


Subject(s)
Anticoagulants , Antithrombin III/pharmacology , Polysaccharides/pharmacology , Xylans/pharmacology , Blood Coagulation Tests , Drug Synergism , Edible Grain/analysis , Factor X/antagonists & inhibitors , Factor Xa , Heparin/pharmacology , Humans , Molecular Weight , Pentosan Sulfuric Polyester/pharmacology , Prothrombin/antagonists & inhibitors , Sulfates/pharmacology , Wood , Xylans/isolation & purification , Zea mays/analysis
19.
Article in English | MEDLINE | ID: mdl-12769657

ABSTRACT

The balance between the activities of the coagulation and fibrinolytic cascades is crucial for normal hemostasis. However, imbalances can lead to pathological thrombotic events, as is observed in heart attacks and strokes, as well as excessive bleeding, as in hemophilia. Recent investigations have uncovered a novel molecular connection between the two cascades that has been termed thrombin-activable fibrinolysis inhibitor (TAFI) as well as procarboxypeptidase U, procarboxypeptidase R or plasma procarboxypeptidase B. TAFI is the precursor of an enzyme (TAFIa) with basic carboxypeptidase activity that attenuates the lysis of fibrin clots by removal of the carboxyl-terminal lysine residues from partially-degraded fibrin that mediate positive feedback in the fibrinolytic cascade. The plasma concentration of TAFI varies substantially (up to approximately 10-fold) in the human population and may constitute a novel risk factor for thrombotic disorders. Sixteen single nucleotide polymorphisms have been identified in the 5'-flanking, protein coding, and 3'-untranslated regions of the TAFI gene. The polymorphisms all have been shown to be associated with variations in plasma TAFI concentrations. One amino acid substitution has been found to directly alter the properties of the TAFIa enzyme. This review provides a general overview of the TAFI pathway, including a discussion of the spectrum of inhibitors of TAFIa that have been described, and summarizes the recent advances in the molecular genetics of the TAFI gene as well as the results of studies that may implicate the TAFI pathway in risk for arterial and venous thrombotic disorders.


Subject(s)
Carboxypeptidase B2/genetics , Carboxypeptidase B2/metabolism , Thrombosis/enzymology , Thrombosis/genetics , Animals , Carboxypeptidase B2/antagonists & inhibitors , Carboxypeptidase B2/chemistry , Humans , Risk Factors , Thrombosis/drug therapy
20.
Thromb Res ; 47(3): 353-63, 1987 Aug 01.
Article in English | MEDLINE | ID: mdl-3629561

ABSTRACT

A protein inhibitor was isolated from commercial preparations of salmon sperm and its physical and anticoagulant properties were compared with an inhibitor isolated earlier from commercial preparation of bovine testicular hyaluronidase. The inhibitor from bovine source was heat and acid labile and had a molecular weight of approximately equal to 35000 while the one from salmon sperm had a molecular weight of approximately equal to 5700 and was stable to heat and acid. To determine the mechanism of the inhibitory effect, a system of purified components consisting of isolated prothrombin, Factor Xa, Factor Va, Ca++, and vesicles of phosphatidylcholine (PCPS, 25% PS) was used. Included also was dansylarginine N- (3-ethyl-1,5-pentanedidyl) amide (DAPA) which binds newly formed thrombin and yields the time course of prothrombin conversion by virtue of enhanced fluorescence of the DAPA - thrombin complex. The inhibitor of bovine testes was effective only when PCPS was the limiting component suggesting that its action was directed against the phospholipid component of the prothrombinase complex. The inhibitor from salmon sperm was found to lower the rate of conversion of prothrombin to thrombin in an in vitro system where thrombin generation was measured by its action on the chromogenic substrate H-D-Phe-Pip-Arg-pNa (S-2238). It inhibited the conversion of Factor X to Xa and also the the amidolytic cleavage by Factor Xa of chromogenic substrate N-Benz-Ile-Glu-Gly-Arg-pNa (S-2222).


Subject(s)
Anticoagulants , Proteins/pharmacology , Spermatozoa/analysis , Testis/analysis , Animals , Anticoagulants/isolation & purification , Blood Coagulation Tests , Cattle , Hot Temperature , Humans , Hydrogen-Ion Concentration , Male , Molecular Weight , Proteins/isolation & purification , Salmon
SELECTION OF CITATIONS
SEARCH DETAIL