+ All Categories

COLMAN

Date post: 25-Sep-2015
Category:
Upload: albertolpz
View: 212 times
Download: 0 times
Share this document with a friend
Description:
20.Thrombin-Activatable Fibrinolysis Inhibitor AKA Procarboxypeptidase U
Popular Tags:
41
Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi 1 de 41 24/06/2006 01:13 p.m. Editors: Colman, Robert W.; Clowes, Alexander W.; Goldhaber, Samuel Z.; Marder, Victor J.; George, James N. Title: Hemostasis and Thrombosis: Basic Principles and Clinical Practice, 5th Edition Copyright ©2006 Lippincott Williams & Wilkins > Table of Contents > Part I - Basic Principles of Hemostasis and Thrombosis > Section B - Fibrinolysis and its Regulation > Chapter 20 - Thrombin-Activatable Fibrinolysis Inhibitor AKA Procarboxypeptidase U Chapter 20 Thrombin-Activatable Fibrinolysis Inhibitor AKA Procarboxypeptidase U Michael E. Nesheim Judith Leurs Dirk F. Hendriks Thrombin-activatable fibrinolysis inhibitor (TAFI) is a plasma glycoprotein of 401 amino acids that circulates at a concentration of approximately 5.0 µg per mL (1,2). It is the precursor of zinc ion–dependent, carboxypeptidase B–like enzyme, designated TAFIa, that suppresses fibrinolysis. It is activated by proteolysis at the Arg92–Ala93 bond. The amino-terminal portion is an activation fragment, and the carboxy-terminal portion is the enzyme TAFIa. Thrombin and plasmin are capable of activating TAFI, but they are relatively inefficient in doing so. The physiologic activator is thought to be the thrombin–thrombomodulin complex (3). The gene for TAFI is located on chromosome 13 (4). It contains 11 exons and spans approximately 48 kb of genomic DNA and is expressed in the liver (5). As is the case with many complex biologic molecules, TAFI made its existence known in several laboratories through independent and unrelated investigations, each of which resulted in a new name
Transcript
  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    1 de 41 24/06/2006 01:13 p.m.

    Editors: Colman, Robert W.; Clowes, Alexander W.;

    Goldhaber, Samuel Z.; Marder, Victor J.; George, James N.

    Title: Hemostasis and Thrombosis: Basic Principles and

    Clinical Practice, 5th Edition

    Copyright 2006 Lippincott Williams & Wilkins

    > Table of Contents > Part I - Basic Principles of Hemostasis and

    Thrombosis > Section B - Fibrinolysis and its Regulation > Chapter 20 -

    Thrombin-Activatable Fibrinolysis Inhibitor AKA Procarboxypeptidase U

    Chapter 20

    Thrombin-Activatable FibrinolysisInhibitor AKA Procarboxypeptidase

    U

    Michael E. Nesheim

    Judith Leurs

    Dirk F. Hendriks

    Thrombin-activatable fibrinolysis inhibitor (TAFI) is a plasma

    glycoprotein of 401 amino acids that circulates at a concentration

    of approximately 5.0 g per mL (1,2). It is the precursor of zinc

    iondependent, carboxypeptidase Blike enzyme, designated

    TAFIa, that suppresses fibrinolysis. It is activated by proteolysis at

    the Arg92Ala93 bond. The amino-terminal portion is an activation

    fragment, and the carboxy-terminal portion is the enzyme TAFIa.

    Thrombin and plasmin are capable of activating TAFI, but they are

    relatively inefficient in doing so. The physiologic activator is

    thought to be the thrombinthrombomodulin complex (3). The

    gene for TAFI is located on chromosome 13 (4). It contains 11

    exons and spans approximately 48 kb of genomic DNA and is

    expressed in the liver (5).

    As is the case with many complex biologic molecules, TAFI made

    its existence known in several laboratories through independent

    and unrelated investigations, each of which resulted in a new name

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    2 de 41 24/06/2006 01:13 p.m.

    for the previously unknown entity. As a consequence, the protein

    has acquired several monikers, each of which, as learned in

    retrospect, refers to the same entity. Among its names are

    procarboxypeptidase U (6,7), plasma procarboxypeptidase B (1,8),

    procarboxypeptidase R (9), and TAFI (2). The name

    procarboxypeptidase U was assigned because the protein is the

    precursor of an unstable carboxypeptidase Blike enzyme found in

    serum. It was named plasma procarboxypeptidase B because it is

    the precursor of carboxypeptidase Blike enzyme homologous to

    the carboxypeptidase B precursor found in the pancreas. It was

    called procarboxypeptidase R because it has a preference for

    removal of arginine, as apposed to lysine, from the carboxy

    terminus of proteins and peptides. This distinguishes the enzyme

    from the other carboxypeptidase Blike enzyme of plasma, known

    as carboxypeptidase N (6). It was named TAFI because it was

    discovered in a search for an entity that gives rise to an inhibitor

    of fibrinolysis in response to prothrombin activation.

    This chapter provides information regarding the presumed balance

    between the deposition and removal of fibrin and the role of the

    TAFI pathway in it; the activation of TAFI; the mechanisms by

    which TAFIa suppresses fibrinolysis; TAFI and the factor

    XIdependent pathway of coagulation; assays for TAFI and TAFIa;

    the physiologic and pathophysiologic roles of the TAFI pathway;

    and an update on the epidemiology of TAFI. Several recent reviews

    on TAFI are available

    (10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26).

    THE BALANCE BETWEEN FIBRINDEPOSITION AND REMOVALThe vasculature system has within it two powerful, well-regulated

    systems that operate to both stop blood flow at the site of an

    injury and to maintain blood fluidity elsewhere. These systems are

    known, respectively, as the coagulation and fibrinolytic cascades.

    These systems involve plasma proteins, formed elements of blood,

    particularly platelets, and cells lining the blood vessel wall. They

    are latent, and therefore their potential is not obvious without

    overt stimulation. When triggered, however, they can be very

    potent. This point has been demonstrated, for example, in

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    3 de 41 24/06/2006 01:13 p.m.

    chimpanzees that were injected over a 30-second period with trace

    quantities of a combination of blood coagulation factor Xa and

    procoagulant phospholipid vesicles (27). Within 1 minute, all of the

    plasma fibrinogen had been converted to fibrin, and the platelet

    count had dropped to zero. This startling and dramatic coagulation

    response, which might be expected to be fatal, had no long-term

    effects on the animals because it was immediately followed by an

    equally potent fibrinolytic response. Within a minute or two, the

    circulating level of tissue-type plasminogen activator (tPA) had

    risen approximately 800-fold, and all of the fibrin that had been

    deposited within the vascular system was solubilized to fibrin

    degradation products. These experiments demonstrated, in the

    systemic circulation, events that presumably can happen locally

    when required to prevent local blood loss or remove

    inappropriately deposited fibrin.

    A further appreciation of the potential of the coagulation system

    can be gained by considering that thrombin added to plasma at 1

    NIH U per mL will provide a clot in approximately 15 seconds. The

    plasma, however, has in it sufficient prothrombin to generate

    approximately 150 NIH U of thrombin per mL if fully activated.

    Therefore, were the coagulation system to be fully activated

    instantaneously, the blood would be fully gelled within 1 or 2

    seconds, and the rate-limiting step would be the polymerization of

    fibrin. Likewise, plasmin sustained at a level of approximately 2 nM

    will lyse a fibrin clot within approximately 30 minutes. Plasma has

    plasminogen in it at a level of approximately 2,000 nM. Therefore,

    if the plasminogen were instantly turned to plasmin, a clot could

    be expected to be fully solubilized in approximately 2 seconds.

    These considerations suggest that the coagulation and fibrinolytic

    systems are potentially extremely powerful. They also tend to

    rationalize the many levels of control that exist in these systems

    so that they can perform their respective functions without doing

    untoward damage to the host.

    The balance between fibrin deposition and removal is depicted in

    Figure 20-1. In response to vascular injury, the coagulation

    cascade is upregulated to convert prothrombin to thrombin, which

    then converts fibrinogen to fibrin, thereby producing the familiar

    blood clot. In response to fibrin, the fibrinolytic cascade can be

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    4 de 41 24/06/2006 01:13 p.m.

    P.382

    upregulated to convert plasminogen to plasmin, which then digests

    fibrin into soluble fibrin

    degradation products. When these processes are properly

    balanced, the physiologic roles of these systems are realized.

    When they are unbalanced, however, bleeding or thrombosis can

    occur. The systems are regulated at many levels, most of which

    are not depicted in the figure. One very important mode for the

    regulation of coagulation, however, is indicated. It involves the

    protein C pathway, whereby the thrombinthrombomodulin

    complex converts the zymogen protein C to the enzyme, activated

    protein C (APC), which, through a negative feedback loop,

    downregulates thrombin formation (28). Studies with TAFI have

    shown that a similar feedback loop exists on fibrinolytic side of the

    balance (2,3). In this case, the thrombinthrombomodulin complex

    activates the zymogen TAFI to the enzyme, TAFIa, which

    suppresses the fibrinolytic cascade. Because TAFI is activated by

    the thrombinthrombomodulin complex, the TAFI pathway provides

    an explicit molecular connection between the coagulation and

    fibrinolytic cascades, such that activation of the former can

    suppress the activation of the latter.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    5 de 41 24/06/2006 01:13 p.m.

    FIGURE 20-1. The balance between fibrin deposition and

    removal. Prothrombin (II) is activated to thrombin (IIa) by the

    coagulation cascade, and fibrin deposition occurs.

    Subsequently, plasminogen (Plg) is activated to plasmin (PLn)

    by the fibrinolytic cascade, and fibrin is removed. The

    thrombinthrombomodulin complex (IIa-TM) activates protein C

    (PC) and thrombin-activatable fibrinolysis inhibitor (TAFI) to

    the enzymes-activated protein C (APC) and TAFIa, which

    respectively downregulate the coagulation and fibrinolytic

    cascades. The TAFI pathway provides a regulatory link between

    the two cascades such that activation of coagulation

    suppresses fibrinolysis. FGN, fibrinogen; FDP; fibrin

    degradation products.

    The importance of the protein C pathway in the regulation of the

    coagulation cascade has been demonstrated by the existence of

    severe thrombosis in the congenital absence of protein C (28,29)

    and the elevated risk of thrombosis in the condition known as

    activation protein C resistance, associated with factor VLeiden (30).

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    6 de 41 24/06/2006 01:13 p.m.

    Whether defects in TAFI exist and to what extent they might be

    associated with hemostatic abnormalities is not known as yet.

    THE ACTIVATION OFTHROMBIN-ACTIVATABLE FIBRINOLYSISINHIBITORTAFI is activated by proteolytic cleavage at the Arg92Ala93 bond

    (1,2). The enzyme TAFIa is composed of amino acids 93 through

    401 (1). Enzymes known to catalyze this cleavage are thrombin,

    plasmin, and trypsin (1,2). The reactions catalyzed by thrombin

    and plasmin are very inefficient compared to many other reactions

    catalyzed by these enzymes. The efficiency of the reaction

    catalyzed by thrombin, however, is stimulated by a factor of 1,250

    by thrombomodulin (3). This magnitude of increase accomplished

    by thrombomodulin is similar to that obtained in protein C

    activation (31). Heparin stimulates the activation of TAFI by

    plasmin, but not to the same extent as thrombomodulin stimulates

    the thrombin-catalyzed reaction (32). Because thrombomodulin so

    potently stimulates TAFI activation by thrombin, the

    thrombinthrombomodulin complex is thought to be the physiologic

    activator.

    The activation of TAFI is calcium iondependent (33). It shows a

    monophasic dependence on the calcium ion concentration with a

    half maximal effect at a concentration of approximately 0.25 mM.

    This is in sharp contrast to the calcium ion concentration

    dependence of protein C activation, which is biphasic with a peak

    at a calcium ion concentration of approximately 0.25 mM (33,34).

    The kinetics of TAFI activation are consistent with what has been

    referred to as an enzyme-central, parallel assembly model (3,35).

    According to this model, as shown in Figure 20-2, the enzyme

    thrombin can bind to either TAFI or thrombomodulin to form the

    corresponding binary complexes. These can interact further to bind

    the third component (either TAFI or thrombomodulin) to form the

    ternary thrombinthrombomodulinTAFI complex, from which the

    enzyme TAFIa is generated. Three parameters are associated with

    this model: the dissociation constant for the

    thrombinthrombomodulin interaction, the Km for thrombin TAFI

    interaction, and the kcat or turnover number, of the ternary

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    7 de 41 24/06/2006 01:13 p.m.

    P.383

    complex. The three respective parameters were evaluated to be 10

    nM, 1 M, and 1 per second, respectively (3). The Km number is

    high relative to the plasma concentration of TAFI, as is the case

    with protein C activation (31). This implies that the rate of TAFI

    activation in vivo would be proportional to

    the plasma concentration, which has in fact been demonstrated

    (36,37). In addition, the relatively low plasma levels of both

    protein C and TAFI, compared to their Km values, indicates that

    they would not compete appreciably with each other for the

    thrombinthrombomodulin complex, and activation of both would

    occur simultaneously with little if any interference from each other.

    The activation of TAFI has been demonstrated not only with soluble

    thrombomodulin but also with the thrombomodulin found in

    endothelial cells (38,39). Consistent with a relative lack of

    interference with each other, competition for protein C activation

    by TAFI on endothelial cells, although present, was only modest

    even at concentrations of TAFI several times its plasma

    concentration (39). The same was observed regarding the

    inhibition of TAFI activation by protein C (38).

    FIGURE 20-2. A model of the mechanism of activation of

    thrombin-activatable fibrinolysis inhibitor (TAFI) by thrombin

    plus thrombomodulin. This is an enzyme-central, parallel

    assembly model whereby thrombin (IIa) interacts with either

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    8 de 41 24/06/2006 01:13 p.m.

    TAFI or thrombomodulin (TM) to form the corresponding binary

    complexes. These then interact further to form the ternary

    thrombinthrombomodulinTAFI complex from which TAFIa is

    generated. Thrombin alone will catalyze activation of TAFI, but

    thrombomodulin increases the efficiency of the process by a

    factor of 1,250.

    Thrombomodulin has five recognized domains (40). In order, from

    the amino-terminus, they are a lectinlike domain, six tandem

    epidermal growth factor (EGF)-like domains, a chondroitin sulfate

    rich domain, a transmembrane domain, and an intracellular

    domain. The minimal structure required for protein C activation

    comprises the fourth, fifth, and sixth EGF-like domains, plus the

    small peptide that connects epidermal growth factor domains three

    and four (41). This same structure is necessary, but not sufficient,

    for TAFI activation (33,42). In addition, the thirteen residues

    comprising the third disulfide loop of the third epidermal growth

    factor domain are required. Therefore, although the elements of

    thrombomodulin structure required for efficient activation of TAFI

    and protein C are similar, they are not identical. Two amino acid

    residues are particularly intriguing. One is Met388, which is found

    in the small peptide that connects the fourth and fifth epidermal

    growth factor domains. This residue is essential for protein C

    activation, but not for TAFI activation (42). In addition, it can be

    oxidized in the presence of neutrophils (43). When this occurs,

    activity with respect to protein C activation is lost, but activity in

    TAFI activation is retained (33). This, in turn, suggests that in an

    inflammatory milieu, a strong shift in the balance between fibrin

    deposition and removal could occur in favor of thrombosis, because

    the anticoagulant pathway through protein C would be severely

    attenuated, but the antifibrinolytic pathway through TAFI would

    not. The other residue that stands out is Phe376, which is in the

    fourth epidermal growth factor domain. When this residue is

    replaced with alanine, activity in TAFI activation is retained, but

    activity in protein C activation is lost; therefore, Phe376 appears

    very important for protein C, but not for TAFI activation (33).

    Because of the differences in elements of structure of

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    9 de 41 24/06/2006 01:13 p.m.

    thrombomodulin required for the two reactions, the potential exists

    to create recombinant forms of thrombomodulin that are specific

    for either protein C or TAFI activation.

    The elements of thrombin structure required for the two reactions

    are overlapping, but not identical. Alanine-scanning mutagenesis of

    thrombin showed that the activation of both TAFI and protein C

    depends on residues of thrombin that map to exosite I, where

    thrombomodulin is known to bind (44). Other residues, however,

    were identified that are selectively needed for one or the other of

    the two reactions. Therefore, a region below the active site was

    found to be necessary specifically for protein C activation, whereas

    another region above the active site was found to be necessary

    specifically for TAFI activation. In addition, mutagenesis studies in

    which key tryptophan residues of thrombin were exchanged for

    phenylalanine showed that Trp215 of the active site of thrombin is

    very important in protein C, but not TAFI, activation, whereas the

    opposite is true for Trp60d (45).

    The elements of structure of TAFI that confer thrombomodulin

    dependence upon its activation have not been identified to date.

    They, however, apparently do not map to amino acids comprising

    the P6 to P3 positions around the activation cleavage site.

    Mutagenesis studies of this region showed that even when several

    of these residues were replaced with those found around the

    thrombin cleavage site in the chain of fibrinogen, full

    thrombomodulin dependence of TAFI activation was retained, and

    no catalytic efficiency was lost (46). Remarkably, when the

    residues of TAFI around the cleavage site were replaced with those

    around the cleavage site of protein C, the mutant TAFI did not

    express well, and its activation by thrombinthrombomodulin was

    very inefficient.

    MECHANISMS BY WHICH ACTIVETHROMBIN-ACTIVATABLE FIBRINOLYSISINHIBITOR SUPPRESSES FIBRINOLYSISTAFIa is a carboxypeptidase Blike enzyme; that is, it catalyzes

    removal of basic (arginine and lysine) residues from the carboxy

    termini of selected peptides or proteins. This property confers

    upon TAFIa its ability to suppress fibrinolysis. When thrombin

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    10 de 41 24/06/2006 01:13 p.m.

    P.384

    catalyzes the removal of fibrinopeptides A and B from the

    amino-termini of the A and B of fibrinogen E-domains, the

    cleaved fibrinogen monomers associate noncovalently with the

    D-domains of neighboring molecules and the fibrin polymer is

    formed, thereby providing the major proteinaceous component of

    the familiar blood clot (47). The fibrin polymer is further stabilized

    by covalent isopeptide bonds formed between the D-domains of

    adjacent fibrin protomers in the clot. These bonds are formed

    through the action of factor XIIIa.

    In response to fibrin, the vasculature is capable of releasing tPA, a

    serine protease enzyme (48). This enzyme then catalyzes

    activation of glu plasminogen through a single proteolytic cleavage

    to form glu plasmin, also a serine protease. The activation of glu

    plasminogen is stimulated by a factor of approximately 500 by

    fibrin (49,50), an effect that presumably keeps plasminogen

    activation localized to the site of a clot. This stimulation occurs

    through a mechanism in which fibrin acts as a template to bind

    both glu plasminogen and tPA (50). Once formed, plasmin begins

    to digest the clot by catalyzing cleavages after selected arginine

    and lysine residues in the , and chain in regions connecting

    the D- and E-domains of the fibrin protomers (47,51). These

    cleavages expose carboxy-terminal lysine residues in the fibrin

    mesh that provide additional binding sites, especially for glu

    plasminogen. As a consequence, the cofactor activity of fibrin in

    glu plasminogen activation increases by a factor of approximately

    3, and glu plasminogen activation is accelerated

    (52,53). In addition, the partially cleaved fibrin serves as a

    cofactor for a reaction in which a peptide comprising the first 77

    amino acids of glu plasminogen and glu plasmin are liberated by a

    cleavage catalyzed by plasmin. These reactions produce species

    known as lys plasminogen and lys plasmin, respectively. Lys

    plasminogen is a much better substrate for tPA than glu

    plasminogen (approximately 20-fold) (49,50,54). Therefore, (lys)

    plasmin formation is further accelerated. These two phenomena

    (upregulation of the cofactor activity of fibrin and generation of lys

    plasminogen) comprise potent positive feedback steps in the

    process of plasminogen activation. TAFIa interferes with this

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    11 de 41 24/06/2006 01:13 p.m.

    positive feedback by removing the newly exposed carboxy-terminal

    arginine and lysine residues as they appear in fibrin (53). It

    therefore eliminates the positive feedback steps in plasminogen

    activation and slows down the process of fibrinolysis.

    TAFIa also functions by modulating the inhibition of plasmin by

    antiplasmin. Both glu and lys plasmin are very rapidly inhibited by

    antiplasmin, such that free plasmin in plasma has a half-life of

    approximately 0.1 second (55). Fibrin, however, attenuates this

    effect somewhat, such that the rate constant for inhibition of

    plasmin by antiplasmin is reduced approximately threefold by

    intact fibrin. As the fibrin is modified by plasmin, the magnitude of

    this effect increases an additional 10- to 15-fold, such that the

    rate constant for inhibition of plasmin in the presence of

    plasmin-modified fibrin is only approximately 2% to 3% of the

    value found in the absence of fibrin (56,57,58). As a consequence,

    plasmin is highly protected from the inhibitor in the presence of

    fibrin, especially when it has newly exposed carboxy-terminal

    lysine and arginine residues. The net effect of this is to

    substantially raise the steady-state level of plasmin during the

    fibrinolytic process, thereby promoting the rate at which the clot is

    dissolved (57). TAFIa, by removing the carboxy-terminal lysine and

    arginine residues, eliminates most of this protective effect.

    Therefore, in the presence of TAFIa, the steady state level of

    plasmin is considerably lower than it is in the absence of TAFIa,

    and the process of fibrinolysis is prolonged (57).

    Another more subtle effect likely occurs directly in the digestion of

    fibrin by plasmin. Fibrin is solubilized when the , , and chains

    of adjacent protomers within the polymer are cleaved. For adjacent

    D- and E-domains on neighboring protomers to be separated, six

    cleavages must occur at the same location. Exhaustive digestion,

    which includes all such connections, is not necessary for complete

    solubilization, because fibrin degradation products can be released

    as a family of molecules of various molecular weights, the larger

    ones having many D, E connections not severed completely

    (59,60). Therefore, fibrin can be completely solubilized with only a

    fraction of all D, E connections cleaved through. The cleavages

    made by plasmin do not occur randomly, presumably because the

    new carboxy-terminal lysine and or arginine residues that result

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    12 de 41 24/06/2006 01:13 p.m.

    from cleavage at one of the , , or tends to bind plasmin and

    localize it so that the other chains are cleaved at the same D, E

    site (60). As a consequence, relatively few cleavages are needed

    to completely solubilize the fibrin. In the presence of TAFIa,

    however, this retention of plasmin does not occur to the same

    extent, and more cleavages are required to completely dissolve the

    fibrin because they occur randomly throughout the fibrin network.

    This also can prolong the process of fibrinolysis.

    Most of the enzymes of coagulation and fibrinolysis can be

    downregulated by protease inhibitors, especially by antithrombin,

    antiplasmin, and plasminogen activator inhibitor-1 (PAI-1). TAFIa

    is exceptional in this regard in that, to date, no physiologic

    inhibitor of it has been reported. Instead, TAFIa spontaneously

    loses activity, an effect that presumably represents the means by

    which it is physiologically downregulated (6,61). The rate of decay

    is highly dependent on temperature, such that at body temperature

    the TAFIa half-life is approximately 10 minutes, but at room

    temperature it is approximately 2 hours, and at ice temperature it

    is indefinitely stable (61). Because TAFIa is unstable, it suppresses

    fibrinolysis only transiently, an effect that in part determines the

    potency of TAFIa as an antifibrinolytic agent (62). The loss of

    activity occurs in the absence of proteolysis, but once TAFIa loses

    activity, it is susceptible to proteolysis by thrombin or plasmin at

    Arg302 (61,63). Mutagenesis studies have identified a region of

    TAFIa comprising residues 302 to 330 to which the tendency to

    decay can be attributed (61,63). A naturally occurring

    polymorphism is found within this region at residue 325, which

    comprises either a threonine or an isoleucine residue (62,64). The

    latter variant of TAFIa has a half-life that is double that of the

    former; in addition, it is approximately 60% more potent as an

    antifibrinolytic agent (62). Approximately 10% of the population is

    homozygous for the more stable variant and 40% for the less

    stable one (64). Whether pathologic tendencies correlate with one

    or the other is currently under investigation.

    When the time to lyze a clot is measured in vitro at various input

    concentrations of TAFIa, it increases at low concentrations and

    eventually appears to reach a plateau (2,3,61,62). Typically, the

    time to lyze in the plateau is three to four times that observed in

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    13 de 41 24/06/2006 01:13 p.m.

    the absence of TAFIa. The concentration needed to obtain a

    half-maximal prolongation is typically approximately 1 nM, a

    concentration which is only approximately 1% of the plasma

    concentration of the zymogen TAFI. Therefore, although TAFIa

    does not appear to completely eliminate fibrinolysis, even at

    relatively high levels, it is very potent in that only a small fraction

    of available TAFI needs to be activated to have an appreciable

    effect. The magnitude of the maximal prolongation obtained with

    TAFIa is directly proportional to its stability. Variants with a

    relatively short half-life therefore show only a small maximal

    increase, whereas those with a long half-life show the opposite. In

    fact, mutagenesis studies have shown that the maximal

    prolongation is directly proportional to the half-life of the variant

    (61,62). An example is shown in Figure 20-3. This observation had

    proven difficult to rationalize, because the expectation is that a

    relatively short half-life could be offset with an elevated TAFIa

    concentration. Further studies, however, provided an explanation

    for this and disclosed a curious property of the fibrinolytic system

    and its regulation by TAFIa (65,66). The studies showed that the

    TAFIa concentration dependence of lysis prolongation is not best

    represented by a saturating function, but rather by relation,

    whereby the lysis time increases linearly with respect to the TAFIa

    concentration up to some critical value, and then logarithmically

    thereafter. Therefore, the lysis timeversus TAFIa concentration

    relation does not show a true plateau; it gives only a superficial

    impression of a plateau because of the linear-to-logarithmic switch

    in the relation. The explanation for this switch is based on the

    proposition that so long as TAFIa is present at or above some key

    threshold value, fibrin degradation essentially ceases, only to

    begin again once TAFIa decays to a level below the threshold

    value. The time interval over which the TAFIa level stays above the

    threshold is determined by both the initial input concentration of

    TAFIa and its half-life for first-order decay. Therefore, although

    TAFIa might, in principle, totally suppress fibrinolysis, it does not

    appear to do so because it decays. A stable carboxypeptidase

    (pancreatic carboxypeptidase B), however, can virtually stop

    fibrinolysis if present at a sufficiently high level (66). If TAFIa

    were to be generated acutely, and then decay, the effect would be

    to delay, but not eliminate, eventual fibrinolysis. If it were to be

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    14 de 41 24/06/2006 01:13 p.m.

    generated chronically, however, such that it were replenished over

    time, a situation might exist whereby fibrinolysis would be

    eliminated so long as the coagulant stimulus were present.

    Whether this can or does occur in vivo is not known, however. This

    raises the intriguing possibility, although, that under some

    conditions, TAFIa might function as an absolute inhibitor of

    fibrinolysis.

    FIGURE 20-3. Prolongation of fibrinolysis by variants of

    thrombin-activatable fibrinolysis inhibitor (TAFI) with different

    half-lives. The time to lyze a clot is prolonged when TAFIa is

    included. Pseudosaturation occurs in the relation between the

    lysis time and the TAFIa concentration. The maximum

    prolongation depends on the half-life of the TAFIa. The data

    shown in solid circles was obtained with a TAFIa variant having

    threonine at position 325 and a half-life of 10 minutes. The

    data shown in solid squares were obtained with a TAFIa variant

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    15 de 41 24/06/2006 01:13 p.m.

    P.385

    having isoleucine at position 325 and a 20-minute half-life. The

    other data were obtained mixtures of the two. They gave

    maximal lysis times between the extremes, as indicated in the

    insert, where TAFI-TI is the proportion of the sample consisting

    of the Ile325 variant. [From Schneider M, Boffa M, Stewart R,

    et al. Two naturally occurring variants of TAFI (Thr-325 and

    Il-325) differ substantially with respect to thermal stability and

    antifibrinolytic activity of the enzyme. J Biol Chem

    2002;277(2):10211030.]

    The combination of pseudosaturation in the relation between the

    time to lyze a clot and the TAFIa concentration, and a tendency for

    reversible inhibitors of TAFIa to stabilize it, gives rise to a complex

    relation between effects on fibrinolysis and the concentration of

    such inhibitors. Therefore, when reversible inhibitors of TAFIa were

    examined in vitro for their effects on the time to lyze a clot, they

    both prolonged and promoted lysis, depending on the dose

    (67,68). Typically, at relatively low concentrations, such inhibitors

    actually retard fibrinolysis, sometimes by a considerable margin,

    because they stabilize the TAFIa population but do not completely

    inhibit all the TAFIa molecules. Only at relatively high

    concentrations are they able to sufficiently inhibit the whole

    population to overcome the stabilizing effect.

    THROMBIN-ACTIVATABLE FIBRINOLYSISINHIBITOR AND THE FACTORXIDEPENDENT PATHWAY OFCOAGULATIONWhen clotting is triggered in whole blood or plasma, a series of

    events occur that collectively have been designated, in sequence,

    the initiation, propagation, and termination phases (69). In the

    initiation phase, events such as platelet activation, factor V and

    factor VIII activation, and prothrombin activation at a low level

    occur. At the end of the initiation phase, clotting occurs. At this

    point, approximately 1% or 2% of the prothrombin has been

    activated. This is followed by the propagation phase in which the

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    16 de 41 24/06/2006 01:13 p.m.

    P.386

    factor XIdependent (intrinsic) pathway is triggered, presumably

    through activation of factor XI by thrombin (70), and massive

    prothrombin activation occurs within the clot. This is followed by

    the termination phase in which the reactions of the coagulation

    cascade subside and thrombin is consumed by antithrombin.

    Samples of plasma with defects in the factor XIdependent

    pathway, such as those with severe hemophilia A or B, display the

    initiation phase (69,71), but not the intense thrombin formation of

    the propagation phase.

    Several investigators noted early on that when clotting and

    subsequent fibrinolysis were induced by adding tPA and thrombin

    to normal plasmas, or those with defects in the factor

    XIdependent pathway, fibrinolysis occurred early in the defective

    plasmas (72,73,74). For example, clots made in normal plasma

    would lyze under the extant condition in 2 hours, and those made

    in the deficient plasmas would lyze in approximately 30 minutes.

    The phenomenon was designated premature lysis (72). The

    mechanism for this subsequently was shown to be dependent on

    the TAFI pathway, in that normal plasma showed premature lysis

    when TAFIa was inhibited, and normal lysis could be restored in

    hemophilia plasma by promoting TAFI activation (72,74). These

    and other studies showed that the massive level of thrombin

    transiently formed after clotting in normal plasma is sufficient,

    even in the absence of thrombomodulin, to activate enough of the

    TAFI pool to subsequently suppress fibrinolysis (71,72,73,74,75).

    This suggests the concept that a role of the factor

    XIdependent pathway of coagulation is to suppress fibrinolysis

    through the activation of TAFI, thereby stabilizing the newly

    formed clot. The concept of premature lysis has also led to the

    hypothesis that bleeding in hemophiliacs is caused as much by a

    failure to trigger the TAFI pathway and therefore suppress

    fibrinolysis as it is by the formation of a clot in the first place. This

    hypothesis, although very plausible, has yet to be tested in a

    systematic way.

    ASSAYS FOR THROMBIN-ACTIVATABLEFIBRINOLYSIS INHIBITOR AND ACTIVE

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    17 de 41 24/06/2006 01:13 p.m.

    THROMBIN-ACTIVATABLE FIBRINOLYSISINHIBITORNumerous assays for TAFI and TAFIa have been described, and

    some are available commercially. The assays for TAFI are either

    immunologic assays based on measurements of antigen or

    functional assays based on measurements of the activity of TAFIa

    following the complete activation of TAFI by

    thrombinthrombomodulin. Some of the immunologic assays are

    compromised somewhat because they respond differentially to

    various forms of the antigen that can arise because of proteolysis

    of it (76,77). In addition, some assays have been shown to

    respond very differently to the Ile325/Thr325 isoforms of TAFI

    (78). Such assays have been applied to the determination of the

    average TAFI concentration and its distribution about the average

    in several large populations. Substantial difference in the averages

    have been reported by several groups, but all report a fairly broad

    concentration distribution (77,79). The individual variations have

    been reported to be determined mostly by genetics, as opposed to

    by environment (80). The difference in average values reported by

    different groups may reflect differences in concentrations assigned

    to assay standards rather than real differences between the

    populations studied.

    Assays for the enzyme TAFIa are based on measuring its

    carboxypeptidase Blike function with a variety of substrates.

    These assays are complicated by the existence at relatively high

    levels of the constitutively active carboxypeptidase Blike enzyme,

    known as carboxypeptidase N, in plasma. Its concentration is

    approximately 100 nM, which is about 100 times the level at which

    TAFIa would have a significant effect on fibrinolysis. Therefore,

    detecting TAFIa at levels in the range of 1 nM, for example,

    requires a substrate that is highly selective for TAFIa or an

    inhibitor that is highly specific for carboxypeptidase N. No

    synthetic substances with absolute specificity have been described

    to date, but the judicious use of partially selective substrates has

    indicated that the endogenous basal level of TAFIa is less than 100

    pM (81,82). Another assay has been described for TAFIa that is on

    the basis of its ability to downregulate plasminogen activation

    (83). In this assay, high-molecular-weight soluble fibrin

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    18 de 41 24/06/2006 01:13 p.m.

    degradation products are incubated for a designated period with

    the plasmin sample containing TAFIa. Following this, the residual

    cofactor activity of the fibrin degradation products is measured in

    cleavage of a fluorescent plasminogen derivative by the vampire

    bat plasminogen activator. This assay is very specific for TAFIa, as

    opposed to carboxypeptidase N, and it responds to TAFIa in the

    sample at levels ranging from 5 to 200 pM. Therefore, it is both

    very specific and highly sensitive. The application of it to five

    freshly drawn plasma samples from apparently healthy volunteers

    showed the basal level of plasma TAFIa to be 11 pM. This is only

    approximately 0.01% of the TAFI level in plasma, suggesting very

    little systemic activation of the TAFI pathway under basal

    conditions.

    ACTIVATION OF THROMBIN-ACTIVATABLEFIBRINOLYSIS INHIBITOR IN VITRO ANDIN VIVOIndirect evidence for the activation of TAFI upon clotting in vitro is

    provided by the timing of subsequent fibrinolysis when a

    plasminogen activator is included in the experiments. The time to

    achieve lysis after clotting is considerably reduced when a

    carboxypeptidase B inhibitor is included. Quantitatively similar

    results are obtained if a monoclonal antibody directed at TAFI that

    prevents its activation is included. From such observations, the

    conclusion is reached that TAFI activation occurs following clotting

    in plasma. Studies to directly measure TAFIa over time, when a

    clot is formed through the coagulation cascade and subsequently

    lyzed because of included tPA, have shown that TAFIa is formed

    shortly after clotting. Its concentration exhibits a transient peak

    that decays with a half-life of approximately 10 minutes. The

    activator is presumably thrombin, formed after the clot is made.

    Some time later, fibrinolysis occurs, and this is accompanied by a

    second transient burst of TAFIa activity; in this case, the activator

    is presumably plasmin (81). TAFIa generated in the first peak

    appears to delay fibrinolysis, but that which occurs in the second

    peak does not because it is likely formed too late in the sequence

    of events. Evidence for activation of TAFI in spontaneously clotting

    whole blood is provided by the transient increase in

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    19 de 41 24/06/2006 01:13 p.m.

    carboxypeptidase Blike activity in serum. This activity is unstable

    and therefore appears only transiently (6). Its appearance, in

    retrospect, provided the first clue to the existence of TAFI. Studies

    in thrombolysis models in animals indirectly indicate that TAFI can

    be activated in vivo and that TAFIa can retard fibrinolysis

    (76,84,85). A particularly revealing study (76) within an arterial

    thrombolysis model in the rabbit showed that a TAFIa inhibitor

    included along with tPA could increase the apparent potency of the

    activator by approximately threefold. It could also reduce the time

    to reperfusion and markedly enhance patency, with no appreciable

    increase in bleeding. A similar study in a dog model showed

    directly that TAFI is activated during thrombolysis and that this

    could be diminished or eliminated with a reversible synthetic

    thrombin inhibitor (86). All of these studies together show that

    TAFI is activated postclotting in vitro and that it can be activated

    in vivo. However, the scope of conditions under which it is

    activated in vivo, the extent to which it is activated, and the

    duration are not yet known in detail.

    PHYSIOLOGIC AND PATHOPHYSIOLOGICROLES OF THE THROMBIN-ACTIVATABLEFIBRINOLYSIS INHIBITOR PATHWAYStudies in vivo and in selected animal models indicate that the

    TAFI pathway suppresses the activity of the fibrinolytic cascade

    when coagulation is triggered. This observation strongly suggests

    that it contributes to the balance between fibrin deposition and

    removal. Because the plasma level of TAFI is considerably lower

    than the Km value for its activation by thrombinthrombomodulin,

    its rate of activation, all other things being equal, would be

    expected to be proportional to its plasma concentration. Therefore,

    the impact on fibrinolysis could be expected to vary with the

    plasma concentration, and this expectation has been confirmed

    experimentally (87). Theoretically, therefore, variations in plasma

    levels would associate with tendencies to bleed or thrombose.

    Whether this occurs has been examined in numerous epidemiologic

    studies

    (88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107

    results of which are summarized in Table 20-1. Among the

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    20 de 41 24/06/2006 01:13 p.m.

    P.387

    P.388

    P.389

    P.390

    P.391

    findings are the following: (a) elevated levels of plasma TAFI are

    found in patients with stroke; (b) men requiring coronary artery

    bypass grafting because of stable angina pectoris had a higher

    plasma TAFI level than age-matched controls; (c) the restenosis

    rate 6 months after percutaneous coronary intervention correlated

    with the plasma TAFI level; (d) increased plasma TAFI correlates

    with an increased incidence of angina pectoris in France but not in

    Northern Ireland; (e) no difference is observed between plasma

    levels of TAFI in those who have a myocardial infarction or

    coronary death compared to controls, but fewer patients than

    controls had a TAFI concentration above the 90th percentile; (f)

    oral contraceptives or hormone replacement therapy are variously

    associated with changes in plasma TAFI levels; (g) plasma TAFI is

    low in patients with liver cirrhosis or dengue hemorrhagic fever;

    and (h) in promyleocytic leukemia, plasma TAFI measured as

    antigen is normal but measured as activity is low. Studies to date

    generally suggest association of the TAFI pathway with various

    thrombotic pathologies, but no definitive mechanistic connections

    have yet been identified.

    TABLE 20-1 OVERVIEW OF STUDIES OF proCPU

    (THROMBIN-ACTIVATABLE FIBRINOLYSIS INHIBITOR) AS

    A RISK FACTOR FOR CARDIOVASCULAR DISEASE

    The TAFI knockout mouse is viable and has no obvious thrombotic

    or bleeding phenotype (137). When crossed with a heterozygous

    plasminogen knockout, however, a TAFI-deficient phenotype is

    clearly evident in models involving both clot lysis and leukocyte

    migration in peritoneal inflammation (138). Therefore, in the

    context of the partially plasminogen-deficient mouse, the observed

    phenotypes with respect to TAFIa deficiency are consistent with

    the conclusion that the TAFI pathway modulates fibrinolysis in

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    21 de 41 24/06/2006 01:13 p.m.

    P.392

    vivo. The TAFI knockout mouse has been demonstrated to have

    readily measured deficiencies in wound healing (139,140). This,

    too, could be a consequence of deregulated fibrinolysis, but it also

    might be a consequence of other actions of the TAFI pathway.

    OTHER POTENTIAL FUNCTIONS OF THEACTIVE THROMBIN-ACTIVATABLEFIBRINOLYSIS INHIBITOR PATHWAYThat the TAFI pathway might have functions other than modulation

    of fibrinolysis is plausible because TAFIa is able to target

    molecules other than partially degraded fibrin. Therefore, it might

    function, like many other members of the carboxypeptidase family

    of enzymes, as modulators of other processes.

    Targets of TAFIa other than plasmin-modified fibrin have been

    identified. It is very active toward bradykinin and some

    encephalins, for example (8). It also effectively catalyzes removal

    of carboxy-terminal arginine or lysine residues from peptides

    associated with inflammation, such as the anaphylatoxins C5a, and

    C3a, and thrombin-cleaved osteopontin, which has adhesive and

    cell-signaling functions thought to be important in inflammatory

    responses (141,142,143). A study by Myles et al. (142) suggested

    that the enzyme TAFIa is considerably more efficient than the

    constitutively active plasma enzyme carboxypeptidase N in

    catalyzing cleavage of peptides with sequences based on

    anaphylatoxins, osteopontin, and bradykinin. They also provided

    data that indicated in their experimental animal model that TAFIa

    was more potent than carboxypeptidase N in preventing a

    hypotensive response to bradykinin. They also suggested that

    thrombin could upregulate the proinflammatory properties of

    osteopontin and that subsequent action of TAFIa could

    down-regulate them.

    Clinical evidence for a potential role of the TAFI pathway comes

    from a recent study by Hovinga et al. on the association between a

    functional single-nucleotide dimorphism in the coding region of the

    TAFI gene and outcome (survival or death) in meningococcal

    disease (144). The dimorphism codes for either threonine or

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    22 de 41 24/06/2006 01:13 p.m.

    isoleucine at amino acid 325 in the TAFI protein. The Ile325

    variant of TAFIa is twice as stable and 60% more potent as an

    antifibrinolytic than the Thr325 variant. The genotype of survivors

    and many of their relatives were determined, as were the

    genotypes of relatives of the nonsurvivors. The analysis indicated

    that patients whose parents were carriers of the TAFIa Ile325

    genotype had a 1.6-fold [confidence interval (CI), 0.7 to 3.7]

    higher risk of contracting meningococcal disease and a 3.1-fold

    (CI, 1.0 to 9.5) increased risk of dying from the disease compared

    with all other genotypes. The mechanistic basis for this can only be

    speculated upon at this point, but the observations suggest that

    the TAFI pathway might be significant in the response to sepsis.

    Two recent reviews have been published on the potential

    connections between the TAFI pathway and inflammation

    (145,146). In them, evidence suggesting that TAFI participates in

    crosstalk between coagulation or fibrinolysis and inflammation is

    discussed. However, a definitive understanding of these linkages

    remains to be gathered.

    References

    1. Eaton DL, Malloy BE, Tsai SP, et al. Isolation, molecular

    cloning, and partial characterization of a novel

    carboxypeptidase B from human plasma. J Biol Chem

    1991;266:2183321838.

    2. Bajzar L, Manuel R, Nesheim ME. Purification and

    characterization of TAFI, a thrombin activatable fibrinolysis

    inhibitor. J Biol Chem 1995;270: 1447714484.

    3. Bajzar L, Morser J, Nesheim M. TAFI, or plasma

    procarboxypeptidase B, couples the coagulation and fibrinolytic

    cascades through the thrombin-thrombomodulin complex. J Biol

    Chem 1996;271:1660316608.

    4. Vanhoof G, Wauters J, Schatteman K, et al. The gene for

    human carboxypeptidase U (CPU)a proposed novel regulator

    of plasminogen activationmaps to 13q14.11. Genomics

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    23 de 41 24/06/2006 01:13 p.m.

    1996;38:454455.

    5. Boffa MB, Reid TS, Joo E, et al. Characterization of the gene

    encoding human TAFI (thrombin-activable fibrinolysis inhibitor;

    plasma procarboxypeptidase B). Biochemistry

    1999;38(20):65476558.

    6. Hendriks D, Scharpe S, van Sande M, et al. Characterization

    of a carboxypeptidase in human serum distinct from

    carboxypeptidase N. J Clin Chem Clin Biochem

    1989;27:277285.

    7. Wang W, Hendriks DF, Scharpe SS. Carboxypeptidase U, a

    plasma carboxypeptidase with high affinity for plasminogen. J

    Biol Chem 1994;269: 1593715944.

    8. Tan AK, Eaton DL. Activation and characterization of

    procarboxypeptidase B from human plasma. Biochemistry

    1995;34:58115816.

    9. Campbell W, Okada H. An arginine specific carboxypeptidase

    generated in blood during coagulation or inflammation which is

    unrelated to carboxypeptidase N or its subunits. Biochem

    Biophys Res Commun 1989;162: 933939.

    10. Nesheim M, Wang W, Boffa M, et al. Thrombomodulin and

    TAFI in the molecular link between coagulation and fibrinolysis.

    Thromb Haemost 1997; 78:386391.

    11. Bouma BN, Meijers JC. Role of blood coagulation factor XI in

    downregulation of fibrinolysis. Curr Opin Hematol

    2000;7(5):266272.

    12. Bajzar L. Thrombin activatable fibrinolysis inhibitor and an

    antifibrinolytic pathway. Arterioscler Thromb Vasc Biol

    2000;20(12):25112518.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    24 de 41 24/06/2006 01:13 p.m.

    13. Booth NA. TAFI meets the sticky ends. Thromb Haemost

    2001;85(1):12.

    14. Wu KK, Matijevic-Aleksic N. Thrombomodulin: a linker of

    coagulation and fibrinolysis and predictor of risk of arterial

    thrombosis. Ann Med 2000;32 (Suppl. 1):7377.

    15. Schatteman K, Goossens F, Leurs J, et al. Carboxypeptidase

    U at the interface between coagulation and fibrinolysis. Clin

    Appl Thromb Hemost 2001;7(2): 93101.

    16. Bouma BN, Marx PF, Mosnier LO, et al. Thrombin-activatable

    fibrinolysis inhibitor (TAFI, plasma procarboxypeptidase B,

    procarboxypeptidase R, procarboxypeptidase U). Thromb Res

    2001;101(5):329354.

    17. Nesheim M. Myocardial infarction and the balance between

    fibrin deposition and removal. Ital Heart J 2001;2(9):641645.

    18. Nagashima M, Yin ZF, Broze GJ Jr, et al.

    Thrombin-activatable fibrinolysis inhibitor (TAFI) deficient mice.

    Front Biosci 2002;7:556568.

    19. Boffa MB, Nesheim ME, Koschinsky ML. Thrombin activable

    fibrinolysis inhibitor (TAFI): molecular genetics of an emerging

    potential risk factor for thrombotic disorders. Curr Drug Targets

    Cardiovasc Haematol Disord 2001; 1(2):5974.

    20. Bouma BN, Meijers JC. Thrombin-activatable fibrinolysis

    inhibitor (TAFI, plasma procarboxypeptidase B,

    procarboxypeptidase R, procarboxypeptidase U). J Thromb

    Haemost 2003;1(7):15661574.

    21. Nesheim M. Thrombin and fibrinolysis. Chest

    2003;124(Suppl. 3):33S39S.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    25 de 41 24/06/2006 01:13 p.m.

    22. Antovic JP. Thrombin activatable fibrinolysis inhibitor

    (TAFI). A link between coagulation and fibrinolysis. Clin Lab

    2003;49(9-10):475486.

    23. Bouma BN, Meijers JC. New insights into factors affecting

    clot stability: a role for thrombin activatable fibrinolysis

    inhibitor (TAFI; plasma procarboxypeptidase B, plasma

    procarboxypeptidase U, procarboxypeptidase R). Semin Hematol

    2004;41(1 Suppl. 1):1319.

    24. Van de Wouwer M, Conway EM. Novel functions of

    thrombomodulin in inflammation. Crit Care Med 2004;32(Suppl.

    5):S254S261.

    25. Bajzar L, Jain N, Wang P, et al. Thrombin activatable

    fibrinolysis inhibitor: not just an inhibitor of fibrinolysis. Crit

    Care Med 2004; 32 (Suppl. 5): S320S324.

    26. Marx PF. Thrombin-activatable fibrinolysis inhibitor. Curr

    Med Chem 2004; 11(17):23352348.

    27. Giles AR, Nesheim ME, Herring SW, et al. The fibrinolytic

    potential of the normal primate following the generation of

    thrombin in vivo. Thromb Haemost 1990;63(3):476481.

    28. Esmon CT. Regulation of blood coagulation. Biochem

    Biophys Acta Protein Struct Mol Enzymol

    2000;1477(1-2):349360.

    29. Nizzi FA Jr, Kaplan HS. Protein C and S deficiency. Semin

    Thromb Hemost 1999;25(3):265272.

    30. Nicolaes GA, Dahlback B. Activated protein C resistance

    (FV(Leiden)) and thrombosis: factor V mutations causing

    hypercoagulable states. Hematol Oncol Clin North Am

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    26 de 41 24/06/2006 01:13 p.m.

    2003;17(1):3761.

    31. Le Bonniec BF, MacGillivray RTA, Esmon CT. Thrombin

    Glu-39 restricts the P3 specificity to nonacidic residues. J Biol

    Chem 1991;266: 1379613803.

    32. Mao SS, Cooper CM, Wood T, et al. Characterization of

    plasmin-mediated activation of plasma procarboxypeptidase B.

    Modulation by glycosaminoglycans. J Biol Chem

    1999;274(49):3504635052.

    33. Wang W, Nagashima M, Schneider M, et al. Elements of the

    primary structure of thrombomodulin required for efficient

    thrombin-activable fibrinolysis inhibitor activation. J Biol Chem

    2000;275:2294222947.

    34. Esmon CT, Esmon NL. Protein C activation. Semin Thromb

    Hemost 1984; 10:122130.

    35. Boskovic DS, Giles AR, Nesheim ME. Studies of the role of

    factor Va in the factor Xa-catalyzed activation of prothrombin,

    fragment 1.2-prethrombin-2 and

    dansyl-L-glutamyl-glycyl-L-arginine-meizothrombin in the

    absence of phospholipid. J Biol Chem 1990;265:1049710505.

    36. Mosnier LO, von dem Borne PAKr, Meijers JCM, et al. Plasma

    TAFI levels influence the clot lysis time in healthy individuals in

    the presence of an intact intrinsic pathway of coagulation.

    Thromb Haemost 1998;80:829839.

    37. Strmqvist M, Schatteman K, Leurs J, et al. Immunological

    assay for the determination of procarboxypeptidase U antigen

    levels in human plasma. Thromb Haemost 2001;85:1217.

    38. Bajzar L, Nesheim ME, Morser J, et al. Both cellular and

    soluble forms of thrombomodulin inhibit fibrinolysis by

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    27 de 41 24/06/2006 01:13 p.m.

    potentiating the activation of thrombin-activable fibrinolysis

    inhibitor. J Biol Chem 1998;273:27922798.

    39. Kokame K, Zheng X, Sadler JE. Activation of thrombin

    activable fibrinolysis inhibitor requires epidermal growth

    factor-like domain 3 of thrombomodulin and is inhibited

    competitively by protein C. J Biol Chem 1998;

    273(20):1213512139.

    40. Sadler JE. Thrombomodulin structure and function. Thromb

    Haemost 1997;78(1):392395.

    41. Clarke JH, Light DR, Blasko E, et al. The short loop between

    epidermal growth factor-like domains 4 and 5 is critical for

    human thrombomodulin function. J Biol Chem

    1993;268:63096315.

    42. Wang W, Nagashima M, Morser J, et al. Comparison of the

    structures of thrombomodulin required for the activation of

    protein C and TAFI. Fibrinolysis and Proteolysis 1998;12:1126.

    43. Glaser CB, Morser J, Clarke JH, et al. Oxidation of a specific

    methionine in thrombomodulin by activated neutrophil products

    blocks cofactor activity. A potential rapid mechanism for

    modulation of coagulation. J Clin Invest 1992;90:25652573.

    44. Hall SC, Nagashima M, Zhao L, et al. Thrombin interacts

    with thrombomodulin, protein C and thrombin activatable

    fibrinlysis inhibitor via specific and distinct domains. J Biol

    Chem 1999;274(36):2551025516.

    45. Bell R, Stevens WK, Jia ZC, et al. Fluorescence properties

    and functional roles of tryptophan residues 60d, 96, 148, 207,

    and 215 of thrombin. J Biol Chem 2000;275(38):2951329520.

    46. Schneider M, Nagashima M, Knappe S, et al. Amino acid

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    28 de 41 24/06/2006 01:13 p.m.

    P.393

    residues in the P6-P3 region of thrombin-activable fibrinolysis

    inhibitor (TAFI) do not determine the thrombomodulin

    dependence of TAFI activation. J Biol Chem

    2002;277(12):99449951.

    47. Doolittle RF. Fibrinogen and fibrin. Ann Rev Biochem

    1984;53:195229.

    48. Wall U, Jern C, Jern S. High capacity for tissue-type

    plasminogen activator release from vascular endothelium in

    vivo. J Hypertens 1997;15:16411647.

    49. Hoylaerts M, Rijken DC, Lijnen HR, et al. Kinetics of the

    activation of plasminogen by human tissue plasminogen

    activator role of fibrin. J Biol Chem 1982;257:29122919.

    50. Horrevoets AJG, Pannekoek H, Nesheim ME. A steady-state

    template model that describes the kinetics of fibrin-stimulated

    Glu1 and Lys78- plasminogen activation by native tissue type

    plasminogen activator and variants that lack either the finger or

    kringle 2 domain. J Biol Chem 1997;272:21832191.

    51. Castellino FJ. Recent advances in the chemistry of the

    fibrinolytic system. Chem Rev 1981;81:431446.

    52. Suenson E, Lutzen O, Thorsen S. Initial

    plasmin-degradation of fibrin as the basis of a positive

    feed-back mechanism in fibrinolysis. Eur J Biochem 1984;

    140:513522.

    53. Wang W, Boffa MB, Bajzar L, et al. A study of the

    mechanism of inhibition of fibrinolysis by activated

    thrombin-activable fibrinolysis inhibitor. J Biol Chem

    1998;273(42):2717627181.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    29 de 41 24/06/2006 01:13 p.m.

    54. Collen D, Lijnen HR. Basic and clinical aspects of fibrinolysis

    and thrombolysis. Blood 1991;78:31143124

    55. Lijnen HR, Collen D. Alpha 2-antiplasmin. In: Barrett AJ,

    Salvesen G, eds. Proteinase inhibitors. New York: Elsevier

    Science Publishers BV, 1986:457476.

    56. Lee AYY, Fredenburgh JC, Stewart RJ, et al. Like fibrin,

    (DD)E, the major degradation product of crosslinked fibrin,

    protects plasmin from inhibition by 2-antiplasmin. Thromb

    Haemost 2001;85(3):502508.

    57. Schneider M, Nesheim M. A study of the protection of

    plasmin from antiplasmin inhibition within an intact fibrin clot

    during the course of clot lysis. J Biol Chem

    2004;279(14):1333313339.

    58. Schneider M, Brufatto N, Neill E, et al. Activated

    thrombin-activatable fibrinolysis inhibitor reduces the ability of

    high molecular weight fibrin degradation products to protect

    plasmin from antiplasmin. J Biol Chem

    2004;279(14):1334013345.

    59. Francis CW, Marder VJ. A molecular model of plasmic

    degradation of crosslinked fibrin. Semin Thromb Haemost

    1982;8:2535.

    60. Walker JB, Nesheim ME. The molecular weights, mass

    distribution, chain composition, and structure of soluble fibrin

    degradation products released from a fibrin clot perfused with

    plasmin. J Biol Chem 1999;274(8):52015212.

    61. Boffa MB, Bell R, Stevens WK, et al. Roles of thermal

    instability and proteolytic cleavage in regulation of activated

    thrombin activable fibrinolysis inhibitor. J Biol Chem

    2000;275(17):1286812878.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    30 de 41 24/06/2006 01:13 p.m.

    62. Schneider M, Boffa M, Stewart R, et al. Two naturally

    occurring variants of TAFI (Thr-325 and Il-325) differ

    substantially with respect to thermal stability and

    antifibrinolytic activity of the enzyme. J Biol Chem

    2002;277(2):10211030.

    63. Marx PF, Hackeng TM, Dawson PE, et al. Inactivation of

    active thrombin-activable fibrinolysis inhibitor takes place by a

    process that involves conformational instability rather than

    proteolytic cleavage. J Biol Chem 2000;275 (17):1241012415.

    64. Brouwers GJ, Vos HL, Leebeek FWG, et al. A novel, possibly

    functional, single nucleotide polymorphism in the coding region

    of the Thrombin-activatable Fibrinolysis Inhibitor (TAFI) gene is

    also associated with TAFI levels. Blood 2001;98(6):19921993.

    65. Leurs J, Nerme V, Sim Y, et al. Carboxypeptidase U (TAFIa)

    prevents lysis from proceeding into the propagation phase

    through a threshold-dependent mechanism. J Thromb Haemost

    2004;2(3):416423.

    66. Walker JB, Bajzar L. The intrinsic threshold of the

    fibrinolytic system is modulated by basic carboxypeptidases,

    but the magnitude of the antifibrinolytic effect of activated

    thrombin-activable fibrinolysis inhibitor is masked by its

    instability. J Biol Chem 2004;279(27):2789627904.

    67. Walker JB, Hughes B, James I, et al. Stabilization versus

    inhibition of TAFIa by competitive inhibitors in vitro. J Biol

    Chem 2003;278(11):89138921.

    68. Schneider M, Nesheim M. Reversible inhibitors of TAFIa can

    both promote and inhibit fibrinolysis. J Thromb Haemost

    2003;1(1):147154.

    69. Brummel KE, Paradis SG, Butenas S, et al. Thrombin

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    31 de 41 24/06/2006 01:13 p.m.

    functions during tissue factor-induced blood coagulation. Blood

    2002;100(1):148152.

    70. Broze GJ Jr, Gailani D. The role of factor XI in coagulation.

    Thromb Haemost 1993;70:7274.

    71. Mann KG, Butenas S, Brummel K. The dynamics of thrombin

    formation. Arterioscler Thromb Vasc Biol 2003;23(1):1725.

    72. Broze GJ Jr, Higuchi DA. Coagulation-dependent inhibition

    of fibrinolysis: role of carboxypeptidase-U and the premature

    lysis of clots from hemophilic plasma. Blood

    1996;88:38153823.

    73. von dem Borne PAKr, Meijers JCM, Bouma BN. Feedback

    activation of factor XI by thrombin in plasma results in

    additional formation of thrombin that protects fibrin clots from

    fibrinolysis. Blood 1995;86:30353042.

    74. von dem Borne PAKr, Bajzar L, Meijers JCM, et al. Thrombin

    mediated activation of factor XI results in a

    thrombin-activatable fibrinolysis inhibitor-dependent inhibition

    of fibrinolysis. J Clin Invest 1997;99(10): 23232327.

    75. Bajzar L, Nesheim ME, Tracy PB. The profibrinolytic effect of

    activated protein C in clots formed from plasma is

    TAFI-dependent. Blood 1996;88: 20932100.

    76. Klement P, Liao P, Bajzar L. A novel approach to arterial

    thrombolysis. Blood 1999;94(8):27352743.

    77. Guimaraes AH, van Tilburg NH, Vos HL, et al. Association

    between thrombin activatable fibrinolysis inhibitor genotype and

    levels in plasma: comparison of different assays. Br J Haematol

    2004;124(5):659665.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    32 de 41 24/06/2006 01:13 p.m.

    78. Gils A, Alessi MC, Brouwers E, et al. Development of a

    genotype 325-specific proCPU/TAFI ELISA. Arterioscler Thromb

    Vasc Biol 2003; 23:11221127.

    79. Chetaille P, Alessi MC, Kouassi D, et al. Plasma TAFI

    antigen variations in healthy subjects. Thromb Haemost

    2000;83(6):902905.

    80. Henry M, Aubert H, Morange PE, et al. Identification of

    polymorphisms in the promoter and the 3 region of the TAFI

    gene: evidence that plasma TAFI antigen levels are strongly

    genetically controlled. Blood 2001;97(7): 20532058.

    81. Leurs J, Wissing BM, Nerme V, et al. Different mechanisms

    contribute to the biphasic pattern of carboxypeptidase U

    (TAFIa) generation during in vitro clot lysis in human plasma.

    Thromb Haemost 2003;89(2):264271.

    82. Mao SS, Colussi D, Bailey CM, et al.

    Electrochemiluminescence assay for basic carboxypeptidases:

    inhibition of basic carboxypeptidases and activation of

    thrombin-activatable fibrinolysis inhibitor. Anal Biochem

    2003;319(1): 159170.

    83. Neill EK, Stewart RJ, Schneider MM, et al. A functional

    assay for measuring activated thrombin-activatable fibrinolysis

    inhibitor in plasma. Anal Biochem 2004;330(2):332341.

    84. Redlitz A, Nicolini FA, Malycky JL, et al. Inducible

    carboxypeptidase activity. A role in clot lysis in vivo. Circulation

    1996;93:13281330.

    85. Refino CJ, DeGuzman L, Schmitt D, et al. Consequences of

    inhibition of plasma carboxypeptidase B on in vivo

    thrombolysis, thrombosis and hemostasis. Fibrinolysis and

    Proteolysis 2000;14(5):305314.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    33 de 41 24/06/2006 01:13 p.m.

    86. Mattsson C, Bjorkman JA, Abrahamsson T, et al. Local

    proCPU (TAFI) activation during thrombolytic treatment in a dog

    model of coronary artery thrombosis can be inhibited with a

    direct, small molecule thrombin inhibitor (melagatran). Thromb

    Haemost 2002;(4):557562.

    87. Mosnier LO, von dem Borne PAKr, Meijers JCM, et al. Plasma

    TAFI levels influence the clot lysis time in healthy individuals in

    the presence of an intact intrinsic pathway of coagulation.

    Thromb Haemost 1998;80:829839.

    88. Libourel E, Bank I, Meinardi J, et al. Co-segregation of

    thrombophilic disorders in factor V leiden carriers; the

    contributions of factor VIII, factor XI, thrombin activatable

    fibrinolysis inhibitor and lipoprotein(a) to the absolute risk of

    venous thromboembolism. Haematologica 2002;87:10681073.

    89. Schroeder V, Kucher N, Kohler HP. Role of thrombin

    activatable fibrinolysis inhibitor (TAFI) in patients with acute

    pulmonary embolism. J Thromb Haemost 2003;1:492493.

    90. van Tilburg NH, Rosendaal FR, Bertina RM. Thrombin

    activatable fibrinolysis inhibitor and the risk for deep vein

    thrombosis. Blood 2000;95:28552859.

    91. Brouwers GJ, Leebeek F, Tanck M, et al. Association

    between thrombinactivatable fibrinolysis inhibitor (TAFI) and

    clinical outcome in patients with unstable angina pectoris.

    Thromb Haemost 2003;90:92100.

    92. Juhan-Vague I, Morange PE, Aubert H, et al. Plasma

    thrombin-activatable fibrinolysis inhibitor antigen concentration

    and genotype in relation to myocardial infarction in the north

    and south of Europe. Arterioscler Thromb Vasc Biol

    2002;22:867873.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    34 de 41 24/06/2006 01:13 p.m.

    93. Juhan-Vague I, Morange PE. Very high TAFI antigen levels

    are associated with a lower risk of hard coronary events: the

    PRIME Study. J Thromb Haemost 2003;1:22432244.

    94. Lau HK, Segev A, Hegele RA, et al. Thrombin-activatable

    fibrinolysis inhibitor (TAFI): a novel predictor of angiographic

    coronary restenosis. Thromb Haemost 2003;90:11871191.

    95. Schatteman K, Goossens F, Leurs J, et al. Fast

    homogeneous assay for plasma procarboxypeptidase U. Clin

    Chem Lab Med 2001;39:806810.

    96. Montaner J, Ribo M, Monasterio J, et al. Thrombin-activable

    fibrinolysis inhibitor levels in the acute phase of ischemic

    stroke. Stroke 2003;34:10381040.

    97. Morange PE, Juhan-Vague I, Scarabin PY, et al. Association

    between TAFI antigen and ala 147Thr polymorphism of the TAFI

    gene and the angina pectoris incidencethe PRIME Study.

    Thromb Haemost 2003;89:554560.

    98. Salomon O, Steinberg DM, Dardik R, et al. Inherited factor

    XI deficiency confers no protection against acute myocardial

    infarction. J Thromb Haemost 2003;1:658661.

    99. Santamaria A, Oliver A, Borrell M, et al. Risk of ischemic

    stroke associated with functional thrombin-activatable

    fibrinolysis inhibitor plasma levels. Stroke 2003;34:23872391.

    100. Schroeder V, Chatterjee T, Mehta H, et al. Thrombin

    activatable fibrinolysis inhibitor (TAFI) levels in patients with

    coronary artery disease investigated by angiography. Thromb

    Haemost 2002;88:10201025.

    101. Silveira A, Schatteman K, Goossens F, et al. Plasma

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    35 de 41 24/06/2006 01:13 p.m.

    P.394

    procarboxypeptidase U in men with symptomatic coronary

    artery disease. Thromb Haemost 2000; 84:364.

    102. Zorio E, Castello R, Falcb C, et al. Thrombin-activatable

    fibrinolysis inhibitor in young patients with myocardial

    infarction and its relationship with the fibrinolytic function and

    the protein C system. Br J Haematol 2003;122: 958965.

    103. Colucci M, Binetti BM, Branca MG, et al. Deficiency of

    thrombin activatable fibrinolysis inhibitor in cirrhosis is

    associated with increased plasma fibrinolysis. Hepatology

    2003;38:230237.

    104. Guo X, Morioka A, Kaneko Y, et al. Arginine

    carboxypeptidase (CPR) in human plasma determined with

    sandwich ELISA. Microbiol Immunol 1999; 43:691698.

    105. Lisman T, Leebeek F, Mosnier L, et al.

    Thrombin-activatable fibrinolysis inhibitor deficiency in cirrhosis

    is not associated with increased plasma fibrinolysis.

    Gastroenterology 2001;121:131139.

    106. van Gorp E, Minnema M, Suharti C, et al. Activation of

    coagulation factor XI, without detectable contact activation in

    dengue haemorrhagic fever. Br J Haematol 2001;113:9499.

    107. van Thiel DH, George M, Fareed J. Low levels of thrombin

    activatable fibrinolysis inhibitor (TAFI) in patients with chronic

    liver disease. Thromb Haemost 2001;85:667670.

    108. Watanabe R, Wada H, Watanabe Y, et al. Activity and

    antigen levels of thrombin-activatable fibrinolysis inhibitor in

    plasma of patients with disseminated intravascular coagulation.

    Thromb Res 2001;104:16.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    36 de 41 24/06/2006 01:13 p.m.

    109. Antovic J, Schulman S, Eelde A, et al. Total

    thrombin-activatable fibrinolysis inhibitor (TAFI) antigen and

    pro-TAFI in patients with haemophilia A. Haemophilia

    2001;7:557560.

    110. Meijers JC, Oudijk EJ, Mosnier LO, et al. Reduced activity

    of TAFI (thrombin-activatable fibrinolysis inhibitor) in acute

    promyelocytic leukaemia. Br J Haematol 2000;108:518523.

    111. Brzosko S, Hryszko T, Lebkowska U, et al. Plasma

    tissue-type plasminogen activator, fibrinogen, and time on

    dialysis prior to transplantation are related to carotid intima

    media thickness in renal transplant recipients. Transplant Proc

    2003;35:29312934.

    112. Hryszko T, Malyszko J, Malyszko JS, et al. Patients on

    peritoneal dialysis but not on hemodialysis have elevated

    concentration and activity of thrombin-activatable fibrinolysis

    inhibitor. Thromb Res 2001;104:233238.

    113. Hryszko T, Malyszko J, Malyszko JS, et al. A possible role

    of thrombinactivatable fibrinolysis inhibitor in disturbances of

    fibrinolytic system in renal transplant recipients. Nephrol Dial

    Transplant 2001;16:16921696.

    114. Malyszko J, Malyszko JS, Hryszko T, et al.

    Thrombin-activatable fibrinolysis inhibitor in kidney transplant

    recipient with dyslipidemia. Transplant Proc

    2003;35:22192221.

    115. Malyszko J, Malyszko JS, Mysliwiec M. Markers of

    endothelial cell injury and thrombin activatable fibrinolysis

    inhibitor in nephrotic syndrome. Blood Coagul Fibrinolysis

    2002;13:615621.

    116. Malyszko J, Suchowierska E, Malyszko JS, et al. Some

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    37 de 41 24/06/2006 01:13 p.m.

    aspects of haemostasis in CAPD patients treated with

    erythropoietin. Kidney Blood Press Res 2002;25:240244.

    117. Malyszko J, Malyszko JS, Hryszko T, et al. Simvastatin

    affects TAFI and thrombomodulin in CAPD patients. Thromb

    Haemost 2001;86:930931.

    118. Malyszko J, Malyszko JS, Hryszko T, et al. Some aspects of

    haemostasis in kidney transplant recipients maintained on

    cyclosporine, azathioprine, and prednisone in comparison to

    patients treated with cyclosporine and prednisone. Transplant

    Proc 2003;35:29402942.

    119. Antovic JP, Yngen M, Ostenson CG, et al. Thrombin

    activatable fibrinolysis inhibitor and hemostatic changes in

    patients with type I diabetes mellitus with and without

    microvascular complications. Blood Coagul Fibrinolysis

    2003;14:551556.

    120. Aubert H, Frere C, Aillaud MF, et al. Weak and

    non-independent association between plasma TAFI antigen

    levels and the insulin resistance syndrome. J Thromb Haemost

    2003;1:791797.

    121. Hori Y, Gabazza EC, Yano Y, et al. Insulin resistance is

    associated with increased circulating level of

    thrombin-activatable fibrinolysis inhibitor in type 2 diabetic

    patients. J Clin Endocrinol Metab 2002;87:660665.

    122. Schatteman K, Van Gaal L, Goossens F, et al. Increased

    procarboxypeptidase U levels in type II diabetes may reflect

    decreased firbinolytic rate. Fibrinolysis and Proteolysis

    2000;14:69.

    123. Yano Y, Kitagawa N, Gabazza EC, et al. Increased plasma

    thrombin-activatable fibrinolysis inhibitor levels in

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    38 de 41 24/06/2006 01:13 p.m.

    normotensive type 2 diabetic patients with microalbuminuria. J

    Clin Endocrinol Metab 2003;88:736741.

    124. Antovic JP, Rafik HR, Antovic A, et al. Does thrombin

    activatable fibrinolysis inhibitor (TAFI) contribute to impairment

    of fibrinolysis in patients with preeclampsia and/or intrauterine

    fetal growth retardation? Thromb Haemost 2002;88:644647.

    125. Bladbjerg EM, Madsen JS, Kristensen SR, et al. Effect of

    long-term hormone replacement therapy on tissue factor

    pathway inhibitor and thrombin activatable fibrinolysis inhibitor

    in healthy postmenopausal women: a randomized controlled

    study. J Thromb Haemost 2003;1:12081214.

    126. Juhan-Vague I, Renucci JF, Grimaux M, et al.

    Thrombinactivatable fibrinolysis inhibitor antigen levels and

    cardiovascular risk factors. Arterioscler Thromb Vasc Biol

    2000;20:21562161.

    127. Kemmeren JM, Algra A, Meijers JC, et al. Effect of second-

    and third- generation oral contraceptives on fibrinolysis in the

    absence and presence of the factor V leiden mutation. Blood

    Coagul Fibrinolysis 2002;13:373381.

    128. Meijers J, Middeldorp S, Tekelenburg W, et al. Increased

    fibrinolytic activity during use of oral contraceptives is

    counteracted by an enhanced factor XI-independent down

    regulation of fibrinolysis: a randomized cross-over study of two

    low-dose oral contraceptives. Thromb Haemost 2000;84:914.

    129. Post MS, Hendriks DF, Van Der Mooren MJ, et al. Oral

    oestradiol/trimegestone replacement reduces

    procarboxypeptidase U (TAFI): a randomized,

    placebo-controlled, 12-week study in early postmenopausal

    women. J Intern Med 2002;251:245251.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    39 de 41 24/06/2006 01:13 p.m.

    130. Stromqvist M, Schatteman K, Leurs J, et al. Immunological

    assay for the determination of procarboxypeptidase U antigen

    levels in human plasma. Thromb Haemost 2001;85:1217.

    131. Schatteman K, Goossens F, Scharpe S, et al. Assay of

    procarboxypeptidase U, a novel determinant of the fibrinolytic

    cascade, in human plasma. Clin Chem 1999;45:807813.

    132. Vogelvang T, Leurs J, van der Mooren MJ, et al. Raloxifene

    reduces procarboxypeptidase U, an antifibrinolytic marker. A

    2-year randomised, placebocontrolled study in healthy early

    postmenopausal women. Menopause 2004; 11:110115.

    133. Mousa HA, Downey C, Alfirevic Z, et al. Thrombin

    activatable fibrinolysis inhibitor and its fibrinolytic effect in

    normal pregnancy. Thromb Haemost 2004;92(5):10251031.

    134. Alacacioglu I, Ozcan MA, Alacacioglu A, et al. Plasma

    levels of thrombin activatable fibrinolysis inhibitor in normal

    and preeclamptic pregnant women. Thromb Res

    2004;114:155159.

    135. Santamaria A, Martinez-Rubio A, Borrell M, et al. Risk of

    acute coronary artery disease associated with functional

    thrombin activatable fibrinolysis inhibitor plasma level.

    Haematologica 2004;89:880881.

    136. Lau HK, Segev A, Hegele RA, et al. Thrombin-activatable

    fibrinolysis inhibitor (TAFI): a novel predictor of angiographic

    coronary restenosis. Thromb Haemost 2003;90(6):11871191.

    137. Nagashima M, Yin ZF, Zhao L, et al. Thrombin-activatable

    fibrinolysis inhibitor (TAFI) deficiency is compatible with murine

    life. J Clin Invest 2002; 109(1):101110.

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    40 de 41 24/06/2006 01:13 p.m.

    138. Swaisgood CM, Schmitt D, Eaton D, et al. In vivo

    regulation of plasminogen function by plasma carboxypeptidase

    B. J Clin Invest 2002;110(9): 12751282.

    139. Te Velde EA, Wagenaar GT, Reijerkerk A, et al. Impaired

    healing of cutaneous wounds and colonic anastomoses in mice

    lacking thrombin-activatable fibrinolysis inhibitor. J Thromb

    Haemost 2003;1(10):20872096.

    140. Boffa MB. TAFI and wound healing: closing a knowledge

    gap. J Thromb Haemost 2003;1(10):20752077.

    141. Denhardt DT, Noda M, O'Regan AW, et al. Osteopontin as a

    means to cope with environmental insults: regulatin of

    inflammation, tissue remodeling, and cell survival. J Clin Invest

    2001;107:10551061.

    142. Myles T, Nishimura T, Yun TH, et al. Thrombin activatable

    fibrinolysis inhibitor, a potential regulator of vascular

    inflammation. J Biol Chem 2003; 278(51):5105951067.

    143. Campbell WD, Lazoura E, Okada N, et al. Inactivation of

    C3a and C5a octapeptides by carboxypeptidase R and

    carboxypeptidase N. Microbiol Immunol 2002; 46(2):131134.

    144. Hovinga JAK, Franco RF, Zago MA, et al. A functional

    single nucleotide polymorphism in the thrombin-activatable

    fibrinolysis inhibitor (TAFI) gene associates with outcome of

    meningococcal disease. J Thromb Haemost 2004;2:5457.

    145. Bajzar L, Jain N, Wang P, et al. Thrombin activatable

    fibrinolysis inhibitor: not just an inhibitor of fibrinolysis. Crit

    Care Med 2004;32:S320S324.

    146. Van de Wouwer M, Conway EM. Novel functions of

  • Ovid: Hemostasis and Thrombosis: Basic Principles and Clinical Practice http://gateway.ut.ovid.com/gw1/ovidweb.cgi

    41 de 41 24/06/2006 01:13 p.m.

    thrombinomodulin in inflammation. Crit Care Med

    2004;32:S254S261.


Recommended