Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Arch Dis Child ; 96(2): 164-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21068076

ABSTRACT

BACKGROUND: Increasing numbers of children are being administered warfarin therapy as thromboprophylaxis. Warfarin has a narrow therapeutic window with a target international normalised ratio (INR) of 2-3.5, called the therapeutic range. The length of time a patient's INR remains within the therapeutic range is calculated as 'time in the therapeutic range'. Risk for haemorrhage in children receiving warfarin is 0.5%/patient-year and minor bleeding 2.3%/patient-year, which increases exponentially for INRs >5.0. Practice among non-bleeding adults with INRs ≥5 and ≤9 is to withhold warfarin and allow the INR to return to the therapeutic range. Faster warfarin clearance is correlated with younger age. METHODS AND RESULTS: The study objective was to determine the safety and effectiveness of a conservative approach for management of INRs >5 in children receiving warfarin. Children receiving warfarin with INRs ≥5 had warfarin withheld followed by a next day INR without vitamin K administration. Eighty-nine children (1-16 years) participated in the study with 2353 INRs performed. Twenty-six children had INRs ≥5, 5% of the total performed, with a mean INR of 5.9. The next day repeat mean INR after withholding one dose of warfarin was 3.3 (range 1.2-6.8) with 89% of INRs falling below 5. There were no overt bleeds or symptomatic thrombotic events in the month following the INR >5. Time in the therapeutic range for children with INRs ≥5 was 68%. CONCLUSIONS: Withholding warfarin alone for management of non-bleeding INRs ≥5 and ≤8 appears to be safe and effective.


Subject(s)
Anticoagulants/adverse effects , Blood Coagulation Disorders/chemically induced , Warfarin/adverse effects , Adolescent , Anticoagulants/administration & dosage , Blood Coagulation Disorders/blood , Child , Child, Preschool , Drug Administration Schedule , Drug Monitoring/methods , Humans , Infant , International Normalized Ratio , Prospective Studies , Thrombosis/prevention & control , Treatment Outcome , Warfarin/administration & dosage
2.
Thromb Res ; 126(2): e110-5, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20584541

ABSTRACT

UNLABELLED: Increasing numbers of children require warfarin thromboprophylaxis. Home INR testing by the patient (PST) has revolutionized warfarin management. However, the family/patient must contact the health team for guidance for warfarin dosing. Patient self management(PSM) prepares a patient performing PST to take an active role in warfarin dosing. Adult studies demonstrate that PSM is safe and effective with improved adherence and treatment satisfaction quality of life (QOL). OBJECTIVE: To estimate the safety and efficacy in children performing PSM or PST, to evaluate warfarin dose decision making in PSM, and warfarin related QOL. METHODS: Warfarinized children performing PST for >3m were randomized to PST or PSM. The PSM group underwent warfarin management education and assumed independent warfarin management. INRs were collected for a year prior to and for 1 year of study to determine TTR and warfarin decision making. QOL was assessed through inventory completion and interviews. RESULTS: 28 children were randomized and followed for 12 months. TTR was (83.9% pre/ post), and 77.7% pre to 83.0% post for PST and PSM (p=0.312). Appropriate warfarin decision making was 90% with no major bleeding episodes and no thromboembolic events. PSM was preferred by families. CONCLUSIONS: PSM for children may be a safe and effective management strategy for warfarinized children. Clinical studies with larger sample size are required.


Subject(s)
Anticoagulants/therapeutic use , Heart Diseases/drug therapy , Warfarin/therapeutic use , Adolescent , Adult , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Child , Humans , Infant , Infant, Newborn , International Normalized Ratio , Pilot Projects , Quality of Life , Self Administration , Warfarin/administration & dosage , Warfarin/adverse effects , Young Adult
3.
Thromb Res ; 123(5): 707-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18786700

ABSTRACT

BACKGROUND: Advances in medical and surgical therapy in children have resulted in increased survival in children with primary illnesses. However, thrombosis is a serious complication of this success and results in mortality and morbidity. Prevention or treatment of thrombosis using warfarin is challenging in children due to its narrow therapeutic index and the unique differences in children, including variable nutritional intake and the occurrence of common concomitant viral or bacterial illnesses which alter warfarin metabolism. The variable response to warfarin in children necessitates frequent International Normalized Ratio (INR) monitoring. Education may improve time in therapeutic range (TTR) a measure of warfarin effect, and a surrogate for patient adherence, safety and efficacy. METHODS: The Pediatric Anticoagulation program (Stollery Children's Hospital) developed a novel child-focused educational program KIDCLOT-POC about warfarin therapy and POC-INR meter use. A total of twenty eight children, and their caregivers, participated in KIDCLOT-POC. Questionnaire score comparisons and practical demonstrations assessed the learners' theoretical and practical knowledge of warfarin management. RESULTS: In caregivers, the median pre, post and knowledge retention questionnaire scores were 50 (IQR 27), 93 (IQR 6) (p<0.0001) and 96 (IQR 6) (p<0.0001), respectively. In the 18 children who were >or=6 years of age, post and knowledge retention questionnaire scores were 90 (IQR 16) and 92 (IQR 23) (p=0.44), respectively. The TTR for all children was 81.7% (SD 13.1). CONCLUSIONS: Implementation of KIDCLOT-POC program appears to promote high knowledge development and retention in children and caregivers and high TTR with no adverse events.


Subject(s)
Anticoagulants/therapeutic use , Patient Education as Topic , Warfarin/therapeutic use , Caregivers , Child , Cohort Studies , Humans , International Normalized Ratio , Prospective Studies , Time Factors
4.
Thromb Res ; 123(6): 845-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19038418

ABSTRACT

UNLABELLED: Enoxaparin is a low molecular weight heparin (LMWH) commonly used for thromboprophylaxis children. Enoxaparin dosing is based on patients' weight and results in decimal dosing. Due to the high concentration of enoxaparin the resultant decimal dose makes precise measurement difficult. Dilution is necessary and often results in ten-fold medication administration errors [Ghaleb MA, Barber N, Franklin BD, Yeung VWS, Khaki ZF, Wong ICK. Systematic review of medication errors in pediatric patients. Ann Pharmacother Oct 2006;40(10):1766-76, Raju TN, Kecskes S, Thornton JP, Perry M, Feldman S. Medication errors in neonatal and paediatric intensive-care units. Lancet Aug 12 1989;2(8659):374-6]. Enoxaparin may be administered in whole milligram doses via insulin syringe, where one milligram of enoxaparin equals one unit on the 100 unit graduated insulin syringe. STUDY DESIGN: A retrospective chart review of 514 children. Data was collected on underlying diagnosis, reason for anticoagulation, anti-Xa levels, hemorrhagic events, and medication errors identified. OUTCOME: to determine the occurrence rate of supra-therapeutic anticoagulation as indicated by anti-Xa levels >1.0 u/ml, when enoxaparin doses are rounded up to the whole milligram, and are administered using insulin syringes. The secondary objectives were to determine if the supra-therapeutic anti-Xa levels were associated with hemorrhagic events. To determine if children achieved and maintained therapeutic anti-Xa range using whole milligram dosing and to evaluate the impact of utilizing insulin syringes for administration on reducing dose measurement errors. RESULTS: All 514 patients were prescribed whole milligram enoxaparin dosing, and achieved therapeutic anti-Xa within a mean time of 2 days. No infant or child required decimal doses to achieve therapeutic levels. Five children achieved an initial supra-therapeutic anti-Xa level (1.04 -1.36 U/ml), requiring a single whole milligram dose decrease. There were no associated hemorrhagic events. CONCLUSION: Whole milligram enoxaparin dosing administered via an insulin syringe safely and effectively, achieved therapeutic levels in infants and children. The reduced incidence of enoxaparin dosing errors suggests that whole milligram enoxaparin dosing via an insulin syringe is a method that should be considered for standard of care.


Subject(s)
Anticoagulants/administration & dosage , Enoxaparin/administration & dosage , Medication Errors/prevention & control , Syringes , Adolescent , Anticoagulants/therapeutic use , Child , Child, Preschool , Clinical Protocols , Cohort Studies , Enoxaparin/therapeutic use , Factor Xa Inhibitors , Female , Humans , Infant , Infant, Newborn , Insulin/administration & dosage , Male , Retrospective Studies
5.
J Thromb Haemost ; 6(5): 848-55, 2008 May.
Article in English | MEDLINE | ID: mdl-18221361

ABSTRACT

BACKGROUND: Carboxypeptidase N (CPN) is a constitutively active basic carboxypeptidase sharing specificity with activated thrombin-activable fibrinolysis inhibitor (TAFIa). Generally, CPN is regarded as being non-antifibrinolytic. However, this assumption has not been thoroughly investigated, particularly with respect to long-term antifibrinolysis. In addition, a recent report has shown that plasmin cleavage increases the catalytic activity of CPN. Therefore, we investigated the antifibrinolytic properties of CPN and plasmin-cleaved CPN (CPNc). METHODS: CPN was incubated with plasmin for various periods of time and the prolongation of clot lysis at various concentrations of CPN/CPNc mixture was investigated in TAFI-depleted plasma. CPN cleavage was analyzed by electrophoresis and catalytic activity was determined by monitoring cleavage of the small substrate, FA-Ala-Lys. RESULTS: CPN exhibited antifibrinolytic properties in plasma clot lysis assays when present at supraphysiological concentrations. Depletion of CPN from plasma decreased the lysis time of clots formed from minimally diluted plasma at low tissue-type plasminogen activator (t-PA) concentrations. Plasmin cleavage of CPN markedly increased the antifibrinolytic properties. CPN and CPNc prolonged lysis in a non-saturable, dose-dependent, and t-PA-dependent manner. At sufficient concentration, CPN and CPNc prolonged lysis at least forty-fivefold. CPNc was 700% more antifibrinolytic than CPN but only 7% more active toward FA-Ala-Lys. The active site inhibitor GEMSA eliminated the antifibrinolytic effects of CPN and CPNc. Antifibrinolytic activity correlated with cleavage of active and/or regulatory subunits, presumably generating heterodimeric CPNc. CONCLUSIONS: Limited proteolysis of CPN by plasmin generates an enzyme with greatly increased antifibrinolytic properties. We speculate that (patho)physiological proteolysis of CPN may generate a long-term antifibrinolytic enzyme.


Subject(s)
Fibrinolysin/metabolism , Fibrinolysis , Lysine Carboxypeptidase/metabolism , Antifibrinolytic Agents , Dimerization , Humans , Tissue Plasminogen Activator
6.
J Thromb Haemost ; 5(6): 1257-64, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17389009

ABSTRACT

BACKGROUND: The antifibrinolytic effect of activated thrombin-activatable fibrinolysis inhibitor (TAFIa) and carboxypeptidase B (CPB) displays threshold behavior. When CPB was used to simulate conditions mimicking continuous TAFIa activity, it affected the lysis of plasma clots differently to clots formed from a minimal fibrinolytic system comprising fibrinogen, plasminogen and alpha(2)-antiplasmin. Whereas CPB saturably prolonged clot lysis in the purified system, the effect of CPB did not appear saturable in plasma clots. METHODS: To rationalize this difference, we investigated the effects of alpha(2)-antiplasmin, alpha(2)-macroglobulin, antithrombin and aprotinin on CPB-mediated antifibrinolysis. RESULTS: CPB alone prolonged fibrinolysis in a saturable manner and the efficacy of CPB increased with decreasing tissue-type plasminogen activator (t-PA) concentration. The inhibitors by themselves did not halt fibrinolysis and the potency of each inhibitor in the absence of CPB mirrored their solution-phase plasmin inhibitory potentials: alpha(2)-antiplasmin approximately equal to aprotinin >> alpha(2)-macroglobulin >> antithrombin. With both CPB and inhibitor present, a synergistic effect was observed. The antifibrinolytic sensitivity to CPB was related to the plasmin inhibitory potential of the inhibitor. CONCLUSIONS: Fibrinolysis could be completely inhibited by alpha(2)-antiplasmin, alpha(2)-macroglobulin and antithrombin, but not aprotinin, in the presence of CPB, and occurred only when the irreversible inhibitor or pool of inhibitors were in excess of plasminogen. Western blot analysis indicated that the CPB-mediated shutdown of fibrinolysis was a result of plasminogen consumption prior to clot lysis. The CPB concentration required for fibrinolytic shutdown was dependent on t-PA concentration and the inhibitory potential of the irreversible inhibitor pool.


Subject(s)
Antifibrinolytic Agents/metabolism , Antifibrinolytic Agents/pharmacology , Carboxypeptidase B/metabolism , Carboxypeptidase B/pharmacology , Fibrinolysis/drug effects , Fibrinolysis/physiology , Antithrombins/metabolism , Antithrombins/pharmacology , Aprotinin/metabolism , Aprotinin/pharmacology , Carboxypeptidase B2/metabolism , Carboxypeptidase B2/pharmacology , Drug Synergism , Fibrinolysin/metabolism , Humans , In Vitro Techniques , Kinetics , Tissue Plasminogen Activator/metabolism , Tissue Plasminogen Activator/pharmacology , alpha-2-Antiplasmin/metabolism , alpha-2-Antiplasmin/pharmacology , alpha-Macroglobulins/metabolism , alpha-Macroglobulins/pharmacology
8.
Ann N Y Acad Sci ; 936: 247-60, 2001.
Article in English | MEDLINE | ID: mdl-11460482

ABSTRACT

Fibrin is a cofactor for the formation of plasmin from plasminogen as catalyzed by tissue plasminogen activator. Initial cleavages of fibrin by plasmin upregulates the cofactor activity of fibrin by exposing carboxyl terminal lysine residues. This effect is eliminated by a carboxypeptidase B-like enzyme generated from the precursor, thrombin activatable fibrinolysis inhibitor (TAFI) that is generated by thrombin during the formation of fibrin. Thus, TAFI and its activation to TAFIa create a link between the coagulation and fibrinolytic cascade, such that activation of the former suppresses the latter. Complete solubilization of fibrin results in a family of very large fibrin degradation products. These also have very substantial tissue plasminogen activator cofactor activity that is very highly downregulated by TAFIa.


Subject(s)
Fibrin/metabolism , Plasminogen/metabolism , Carboxypeptidase B2 , Carboxypeptidases/metabolism , Fibrin/chemistry , Fibrin Fibrinogen Degradation Products/metabolism , Hydrolysis , Kinetics
9.
J Biol Chem ; 276(31): 28686-93, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11384970

ABSTRACT

The plasma zymogen prothrombin (II) is converted to the clotting enzyme thrombin (IIa) by two prothrombinase-catalyzed proteolytic cleavages. Thus, two intermediates, meizothrombin (mIIa) and prethrombin-2 (P2), are possible on the reaction pathway. Measurements of the time courses of II, mIIa, P2, and IIa suggested a channeling phenomenon, whereby a portion of the II is converted directly to IIa without free mIIa and P2 as obligatory intermediates. Evidence for this was that the maximum rate of IIa formation preceded the maximum in the level of either intermediate. In addition, analysis of the data according to a model that included two parallel pathways through mIIa and P2 indicated that about 40% of the II consumed did not yield free mIIa or P2. Further studies were carried out in which II was continuously infused in a reactor at a constant rate. Under these conditions II, mIIa, and P2 reached constant steady-state levels, and IIa was produced at a constant rate, equal to that of II infusion. During the steady state, traces of II, mIIa, and P2 were introduced as radiolabels. Time courses of isotope consumption were first order, thus allowing the rates of consumption of II, mIIa, and P2 to be calculated. Under these conditions the rate of II consumption equaled the rate of IIa formation. Rates of consumption of the free intermediates, however, were only 22 (mIIa) and 15% (P2), respectively, of the rate of thrombin formation. Thus, both the time course experiments and the steady-state experiments indicate that an appreciable fraction of II is channeled directly to IIa without proceeding through the free intermediates mIIa and P2.


Subject(s)
Enzyme Precursors/metabolism , Prothrombin/chemistry , Prothrombin/metabolism , Thrombin/metabolism , Animals , Catalysis , Cattle , Chromatography, Affinity , Endopeptidases/metabolism , Enzyme Activation , Factor V/metabolism , Factor X/metabolism , Factor Xa/metabolism , Kinetics , Models, Chemical , Prothrombin/isolation & purification
10.
Arterioscler Thromb Vasc Biol ; 20(12): 2511-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11116046

ABSTRACT

Coagulation and fibrinolysis are processes that form and dissolve fibrin, respectively. These processes are exquisitely regulated and protect the organism from excessive blood loss or excessive fibrin deposition. Regulation of these cascades is accomplished by a variety of mechanisms involving cellular responses, flow, and protein-protein interactions. With respect to regulation mediated by protein-protein interaction, the coagulation cascade appears to be more complex than the fibrinolytic cascade because it has more components. Yet each cascade is regulated by initiators, cofactors, feedback reactions, and inhibitors. Coagulation is also controlled by an anticoagulant pathway composed of (minimally) thrombin, thrombomodulin, and protein C.(1) Protein C is converted by the thrombin/thrombomodulin complex to activated protein C (APC), which catalyzes the proteolytic inactivation of the essential cofactors required for thrombin formation, factors Va and VIIIa. An analogous antifibrinolytic pathway has been identified recently. This pathway provides an apparent symmetry between coagulation and fibrinolysis and is also composed of thrombin, thrombomodulin, and a zymogen that is activated to an enzyme. The enzyme proteolytically inactivates a cofactor to attenuate fibrinolysis. However, unlike APC, which is a serine protease, the antifibrinolytic enzyme is a metalloprotease that exhibits carboxypeptidase B-like activity. Within a few years of each other, 5 groups independently described a molecule that accounts for this antifibrinolytic activity. We refer to this molecule as thrombin activatable fibrinolysis inhibitor (TAFI), a name that is based on functional properties by which it was identified, assayed, and purified. (Because of the preferences of some journals "activatable" is occasionally referred to as "activable.") This review will encompass a historical account of efforts to isolate TAFI and characterize it with respect to its activation, activity, regulation, and potential function in vivo.


Subject(s)
Antifibrinolytic Agents/isolation & purification , Carboxypeptidases/isolation & purification , Fibrinolysis , Thrombin/physiology , Antifibrinolytic Agents/metabolism , Antifibrinolytic Agents/pharmacology , Binding Sites , Carboxypeptidase B2 , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Carboxypeptidases/pharmacology , Cell-Free System , Chromatography, Affinity , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Enzyme Stability , Humans , Isoenzymes/isolation & purification , Kinetics , Protein C/metabolism , Substrate Specificity , Terminology as Topic , Thrombomodulin/metabolism
11.
J Biol Chem ; 275(47): 36612-20, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-10970891

ABSTRACT

A complex of d-dimer noncovalently associated with fragment E ((DD)E), a degradation product of cross-linked fibrin that binds tissue plasminogen activator (t-PA) and plasminogen (Pg) with affinities similar to those of fibrin, compromises the fibrin specificity of t-PA by stimulating systemic Pg activation. In this study, we examined the effect of thrombin-activable fibrinolysis inhibitor (TAFI), a latent carboxypeptidase B (CPB)-like enzyme, on the stimulatory activity of (DD)E. Incubation of (DD)E with activated TAFI (TAFIa) or CPB (a) produces a 96% reduction in the capacity of (DD)E to stimulate t-PA-mediated activation of Glu- or Lys-Pg by reducing k(cat) and increasing K(m) for the reaction; (b) induces the release of 8 mol of lysine/mol of (DD)E, although most of the stimulatory activity is lost after release of only 4 mol of lysine/mol (DD)E; and (c) reduces the affinity of (DD)E for Glu-Pg, Lys-Pg, and t-PA by 2-, 4-, and 160-fold, respectively. Because TAFIa- or CPB-exposed (DD)E produces little stimulation of Glu-Pg activation by t-PA, (DD)E is not degraded into fragment E and d-dimer, the latter of which has been reported to impair fibrin polymerization. These data suggest a novel role for TAFIa. By attenuating systemic Pg activation by (DD)E, TAFIa renders t-PA more fibrin-specific.


Subject(s)
Carboxypeptidases/metabolism , Fibrin Fibrinogen Degradation Products/metabolism , Fibrin/metabolism , Tissue Plasminogen Activator/metabolism , Amino Acid Chloromethyl Ketones/metabolism , Arginine/metabolism , Carboxypeptidase B , Carboxypeptidase B2 , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Fibrinolysin/metabolism , Humans , Kinetics , Lysine/metabolism , Models, Chemical
12.
Blood ; 94(8): 2735-43, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10515877

ABSTRACT

Achieving early, complete, and sustained reperfusion after acute myocardial infarction does not occur in approximately 50% of patients, even with the most potent established thrombolytic therapy. Bleeding is observed with increased concentrations of thrombolytics as well as with adjunctive antithrombotic and antiplatelet agents. A novel approach to enhance thrombolytic therapy is to inhibit the activated form of thrombin-activatable fibrinolysis inhibitor (TAFI), which attenuates fibrinolysis in clots formed from human plasma. Identification of TAFI in rabbit plasma facilitated the development of a rabbit arterial thrombolysis model to compare the thrombolytic efficacy of tissue-plasminogen activator (tPA) alone or with an inhibitor, isolated from the potato tuber (PTI), of activated TAFI (TAFIa). Efficacy was assessed by determining the time to patency, the time the vessel remained patent, the maximal blood flow achieved during therapy, the percentage of the original thrombus, which lysed, the percentage change in clot weight, the net clot accreted, and the release of radioactive fibrin degradation products into the circulation. The results indicate that coadministration of PTI and tPA significantly improved tPA-induced thrombolysis without adversely affecting blood pressure, activated partial thromboplastin time, thrombin clotting time, fibrinogen, or alpha-2-antiplasmin concentrations. The data indicate that inhibitors of TAFIa may comprise novel and very effective adjuncts to tPA and improve thrombolytic therapy to achieve both clot lysis and vessel patency.


Subject(s)
Carboxypeptidases/antagonists & inhibitors , Fibrinolysis/drug effects , Myocardial Infarction/drug therapy , Plant Proteins/therapeutic use , Protease Inhibitors/therapeutic use , Thrombolytic Therapy/methods , Animals , Carboxypeptidase B2 , Carboxypeptidases/blood , Drug Evaluation, Preclinical , Drug Synergism , Drug Therapy, Combination , Fibrin/analysis , Humans , Male , Myocardial Infarction/enzymology , Partial Thromboplastin Time , Plant Proteins/pharmacokinetics , Plant Proteins/pharmacology , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/pharmacology , Rabbits , Specific Pathogen-Free Organisms , Tissue Plasminogen Activator/therapeutic use
13.
Thromb Haemost ; 80(6): 949-55, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9869166

ABSTRACT

Thrombin-activatable fibrinolysis inhibitor (TAFI) is synthesized by the liver and is thought to circulate in plasma as a plasminogen-bound zymogen. When it is activated by the thrombin/thrombomodulin complex, activated TAFI exhibits carboxypeptidase B-like activity. To study the structure-function relationship of TAFI, we expressed recombinant human TAFI in insect cells. During the cloning of TAFI cDNA from several human liver cDNA libraries, we identified a second TAFI cDNA which differed from the published sequence at 2 positions. One of these sequences resulted in a substitution of alanine for threonine at residue 147, the other was a silent mutation. These substitutions were found in several cDNA libraries from different sources. Using Southern blot analysis, we confirmed the existence of this TAFI polymorphism in the population. In order to compare the activation and activity of TAFI isoforms, we expressed both isoforms in the baculovirus expression system, and compared the enzyme kinetics of the purified proteins. The molecular weight of recombinant TAFI is lower than plasma TAFI due to differences in glycosylation. The two recombinant TAFI isoforms had similar activation kinetics and the activated enzymes had similar carboxypeptidase B-like activity towards small molecule substrates. Their ability to retard clot lysis was found to be similar in a plate clot lysis assay.


Subject(s)
Carboxypeptidases/isolation & purification , Protein Isoforms/isolation & purification , Animals , Carboxypeptidase B , Carboxypeptidase B2 , Carboxypeptidases/chemistry , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Cell Line , DNA, Complementary/genetics , Fibrinolysis/drug effects , Genetic Vectors/genetics , Glycosylation , Humans , Kinetics , Molecular Weight , Nucleopolyhedroviruses/genetics , Polymorphism, Genetic , Protein Isoforms/chemistry , Protein Processing, Post-Translational , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Spodoptera/cytology , Spodoptera/metabolism , Structure-Activity Relationship
14.
J Biol Chem ; 273(42): 27176-81, 1998 Oct 16.
Article in English | MEDLINE | ID: mdl-9765237

ABSTRACT

TAFI (thrombin-activable fibrinolysis inhibitor) is a recently described plasma zymogen that, when exposed to the thrombin-thrombomodulin complex, is converted by proteolysis at Arg92 to a basic carboxypeptidase that inhibits fibrinolysis (TAFIa). The studies described here were undertaken to elucidate the molecular basis for the inhibition of fibrinolysis. When TAFIa is included in a clot undergoing fibrinolysis induced by tissue plasminogen activator and plasminogen, the time to achieve lysis is prolonged, and free arginine and lysine are released over time. In addition, TAFIa prevents a 2.5-fold increase in the rate constant for plasminogen activation which occurs when fibrin is modified by plasmin in the early course of fibrin degradation. The effect is specific for the Glu- form of plasminogen. TAFIa prevents or at least attenuates positive feedback expressed through Lys-plasminogen formation during the process of fibrinolysis initiated by tissue plasminogen activator and plasminogen. TAFIa also inhibits plasmin activity in a clot and prolongs fibrinolysis initiated with plasmin. We conclude that TAFIa suppresses fibrinolysis by removing COOH-terminal lysine and arginine residues from fibrin, thereby reducing its cofactor functions in both plasminogen activation and the positive feedback conversion of Glu-plasminogen to Lys-plasminogen. At relatively elevated concentrations, it also directly inhibits plasmin.


Subject(s)
Carboxypeptidases/pharmacology , Enzyme Precursors/pharmacology , Fibrinolysis/drug effects , Arginine/metabolism , Carboxypeptidase B2 , Enzyme Activation , Fibrin/metabolism , Fibrinolysin/antagonists & inhibitors , Humans , Lysine/metabolism , Peptide Fragments/metabolism , Plasminogen/metabolism , Plasminogen Activators
15.
J Biol Chem ; 273(5): 2792-8, 1998 Jan 30.
Article in English | MEDLINE | ID: mdl-9446587

ABSTRACT

Thrombin-activable fibrinolysis inhibitor (TAFI) is a recently described plasma zymogen that can be activated by thrombin to an enzyme with carboxypeptidase B-like activity. The enzyme, TAFIa, potently attentuates fibrinolysis. TAFI activation, like protein C activation, is augmented about 1250-fold by thrombomodulin (TM). In this work, the effects of both soluble and cellular forms of TM on TAFI activation-dependent suppression of fibrinolysis were investigated. Soluble TM included in clots formed from purified components, barium citrate-adsorbed plasma, or normal human plasma maximally increased the tissue plasminogen activator-induced lysis time 2-3-fold, with saturation occurring at 5, 10, and 1 nM TM in the three respective systems. Soluble TM did not effect lysis in the system of purified components lacking TAFI or in plasmas immunodepleted of TAFI. In addition, the antifibrinolytic effect of TM was negated by monoclonal antibodies against either TAFI or TM. The inhibition of fibrinolysis by cellular TM was assessed by forming clots in dialyzed, barium citrate-adsorbed, or normal plasma over cultured human umbilical vein endothelial cells (HUVECs). Tissue plasminogen activator-induced lysis time was increased 2-fold, with both plasmas, in the presence of HUVECs. The antifibrinolytic effect of HUVECs was abolished 66% by specific anti-TAFI or anti-TM monoclonal antibodies. A newly developed functional assay demonstrated that HUVECs potentiate the thrombin-catalyzed, TM-dependent formation of activated TAFI. Thus, endothelial cell TM, in vitro at least, appears to participate in the regulation of not only coagulation but also fibrinolysis.


Subject(s)
Carboxypeptidases/metabolism , Enzyme Precursors/metabolism , Fibrinolysis , Thrombomodulin/metabolism , Blood Proteins/isolation & purification , Blood Proteins/metabolism , Carboxypeptidase B2 , Endothelium, Vascular/physiology , Enzyme Activation , Humans , Protein C/metabolism , Solubility , Thrombin/metabolism
16.
J Biol Chem ; 273(4): 2127-35, 1998 Jan 23.
Article in English | MEDLINE | ID: mdl-9442053

ABSTRACT

Thrombin-activable fibrinolysis inhibitor (TAFI) is a human plasma zymogen similar to pancreatic pro-carboxypeptidase B. Cleavage of the zymogen by thrombin/thrombomodulin generates the enzyme, activated TAFI (TAFIa), which retards fibrin clot lysis in vitro and likely modulates fibrinolysis in vivo. In the present work we stably expressed recombinant TAFI in baby hamster kidney cells, purified it to homogeneity from conditioned serum-free medium, and compared it to plasma TAFI (pTAFI) with respect to glycosylation and kinetics of activation by thrombin/thrombomodulin. Although rTAFI is glycosylated somewhat differently than pTAFI, cleavage products with thrombin/thrombomodulin are indistinguishable, and parameters of activation kinetics are very similar with kcat = 0.55 s-1, K(m) = 0.54 microM, and Kd = 6.0 nM for rTAFI and kcat = 0.61 s-1, K(m) = 0.55 microM, and Kd = 6.6 nM for pTAFI. The respective TAFIa species also were prepared and compared with respect to thermal stability and enzymatic properties, including inhibition of fibrinolysis. The half-life of both enzymes at 37 degrees C is about 10 min, and the decay of enzymatic activity is associated with a quenching (to approximately 62% of the initial value at 60 min) of the intrinsic fluorescence of the enzyme. Stability was highly temperature-dependent, which, according to transition state theory, indicates both high enthalpy and entropy changes associated with inactivation (delta Ho++ approximately equal to 45 kcal/mol and delta So++ approximately equal to 80 cal/mol/K). Both species of TAFIa are stabilized by the competitive inhibitors 2-guanidinoethylmercaptosuccinic acid and epsilon-aminocaproic acid. rTAFIa and pTAFIa are very similar with respect to kinetics of cleavage of small substrates, susceptibility to inhibitors, and ability to retard both tPA-induced and plasmin-mediated fibrinolysis. These studies provide new insights into the thermal instability of TAFIa, a property which could be a significant regulator of its activity in vivo; in addition, they show that rTAFI and rTAFIa are excellent surrogates for the natural plasma-derived species, a necessary prerequisite for future studies of structure and function by site-specific mutagenesis.


Subject(s)
Carboxypeptidases/blood , Thrombin/metabolism , Thrombomodulin/metabolism , Animals , Carboxypeptidase B2 , Carboxypeptidases/chemistry , Cell Line , Cricetinae , Enzyme Activation , Enzyme Stability , Glycosylation , Half-Life , Hot Temperature , Humans , Recombinant Proteins/blood , Transfection
17.
Thromb Haemost ; 78(1): 386-91, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9198184

ABSTRACT

The thrombin thrombomodulin dependent activation of the plasma protein TAFI (Thrombin Activatable Fibrinolysis Inhibitor) and Subsequent Inhibition of Fibrinolysis by the TAFIa is described. Work to date indicates that TAFIa is a carboxypeptidase B enzyme that suppress fibrinolysis most likely by down regulating the cofactor functions of partially degraded fibrin. The existence of TAFI provides the explanation for the apparent profibrinolytic effect of activated protein C. and implies the existence of an explicit molecular connection between the blood coagulation of fibrinolytic cascades that is expressed through the thrombin thrombomodulin dependent activation of TAFI. Thus, thrombin generation can, in principle, result in the suppression of fibrinolysis.


Subject(s)
Blood Coagulation/physiology , Carboxypeptidases/physiology , Fibrinolysis/physiology , Thrombin/physiology , Thrombomodulin/physiology , Amino Acid Sequence , Carboxypeptidase B2 , Carboxypeptidases/isolation & purification , Enzyme Activation , Humans
18.
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
19.
J Biol Chem ; 272(10): 6194-200, 1997 Mar 07.
Article in English | MEDLINE | ID: mdl-9045633

ABSTRACT

Recombinant human prothrombin (rII) and two mutant forms (R155A, R271A,R284A (rMZ) and R271A,R284A (rMZdesF1)) were expressed in mammalian cells. Following activation and purification, recombinant thrombin (rIIa) and stable analogues of meizothrombin (rMZa) and meizothrombin(desF1) (rMZdesF1a) were obtained. Studies of the activation of protein C in the presence of recombinant soluble thrombomodulin (TM) show TM-dependent stimulation of protein C activation by all three enzymes and, in the presence of phosphatidylserine/phosphatidylcholine phospholipid vesicles, rMZa is 6-fold more potent than rIIa. In the presence of TM, rMZa was also shown to be an effective activator of TAFI (thrombin-activatable fibrinolysis inhibitor) (Bajzar, L., Manuel, R., and Nesheim, M. E. (1995) J. Biol. Chem. 270, 14477-14484). All three enzymes were capable of inducing platelet aggregation, but 60-fold higher concentrations of rMZa and rMZdesF1a were required to achieve the effects obtained with rIIa. Second order rate constants (M-1.min-1) for inhibition by antithrombin III (AT-III) were 2.44 x 10(5) (rIIa), 6.10 x 10(4) (rMZa), and 1.05 x 10(5) (rMZdesF1a). The inhibition of rMZa and rMZdesF1a by AT-III is not affected by heparin. All three enzymes bound similarly to hirudin. The results of this and previous studies imply that full-length meizothrombin has marginal procoagulant properties compared to thrombin. However, meizothrombin has potent anticoagulant properties, expressed through TM-dependent activation of protein C, and can contribute to down-regulation of fibrinolysis through the TM-dependent activation of TAFI.


Subject(s)
Enzyme Precursors/physiology , Thrombin/physiology , Antithrombin III/metabolism , Arginine/analogs & derivatives , Arginine/metabolism , Binding, Competitive , Blood Coagulation , Dansyl Compounds/metabolism , Enzyme Activation , Hirudins/metabolism , Humans , Platelet Aggregation , Protein C/metabolism , Recombinant Proteins , Structure-Activity Relationship , Thrombomodulin/metabolism
20.
Blood ; 88(6): 2093-100, 1996 Sep 15.
Article in English | MEDLINE | ID: mdl-8822928

ABSTRACT

Thrombin-activatable fibrinolysis inhibitor (TAFI) is the precursor of an exopeptidase that is identical to plasma procarboxypeptidase B. Upon activation by thrombin, activated TAFI (TAFIa) attenuates fibrinolysis, presumably by catalyzing the removal of C-terminal lysines from partially degraded fibrin. Activated protein C (APC) proteolytically inactivates the essential cofactor in prothrombinase, factor Va, and limits both the formation of thrombin and subsequent activation of TAFI, thereby appearing profibrinolytic. TAFI is able to reconstitute an APC-dependent shortening of lysis time in a purified system; however, it remained to be determined the extent to which TAFI is involved in the profibrinolytic effect of APC in a plasma-based system. To aid in addressing this question, two monoclonal antibodies (MoAbTAFI#16 and #13) and a polyclonal antibody were produced against purified TAFI. MoAbTAFI#16 was shown to inhibit TAFI activation and thereby appears to stimulate fibrinolysis. Furthermore, an enzyme-linked immunosorbent assay was developed using MoAbTAFI#13 and the polyclonal antibody. Through its use, the plasma concentration of TAFI was determined to be 73 nmol/L. In addition, a turbidity assay was used to determine the effect of APC on tissue plasminogen activator-induced fibrinolysis of clots produced from normal human plasma (NHP), plasma immunodepleted of TAFI (TdP), and TdP reconstituted with purified TAFI. APC shortened lysis time of clots produced from NHP in a saturable and concentration-dependent manner. However, APC had no effect on lysis time of clots formed from either TdP or NHP in the presence of 80 nmol/L MoAbTAFI#16. The APC effect could be reconstituted in TdP by the addition of purified TAFI. The lysis time in TdP was increased from 50 to 180 minutes in a TAFI concentration-dependent manner. The EC50 was 15 nmol/L and saturation was approached at physiologically relevant concentrations (60 nmol/L). The profibrinolytic effect of APC was also compared with that of MoAbTAFI#16 and two competitive inhibitors, an inhibitor of the carboxypeptidase A and B family purified from potato tubers and 2-Guanidinoethylmercaptosuccinic acid (GEMSA). All were able to reduce lysis time of clots formed from normal human plasma by 90 minutes, yielding respective EC50 values of 5 nmol/L, 15 nmol/L, 50 nmol/L, and 90 mumol/L. Therefore, the majority of the profibrinolytic effect of APC, in an in vitro plasma system, is dependent on TAFI. Because TAFIa dramatically influences lysis time, inhibitors of TAFIa or TAFI activation may prove to be important adjuvants for thrombolytic therapy.


Subject(s)
Carboxypeptidases/physiology , Fibrinolysis , Protein C/physiology , Antibodies, Monoclonal/immunology , Blood Coagulation , Carboxypeptidase B2 , Enzyme Activation , Humans , Protease Inhibitors/pharmacology , Thrombin/metabolism , Thrombomodulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL