Clin Biochem, Vol. 23, pp. 197-211, 1990 Printed in Canada. All rights reserved.

0009-9120/90 $3.00 + .00 Copyright ¢ 1990 The Canadian Society of Clinical Chemists.

Biochemical and Biological Aspects of the Plasminogen Activation System MICHAEL MAYER Department of Clinical Biochemistry, Hadassah Medical Center and Hadassah-Hebrew University Medical School, P.O. Box 12000, Jerusalem, IL-91120, Israel Plasminogen activators (PAs) are specific proteolytic enzymes which convert the inactive proenzyme plasminogen to plasmin. The plasmin formed is a potent and nonspecific protease which cleaves blood fibrin clots and several other extracellular proteins. In addition to their primary role in the initiation of fibrinolysis, PAs are implicated in a variety of basic biological processes, such as, degradation of the extracellular matrix, tumor invasiveness, tissue remodelling, and cellular differentiation. This review describes recent observations on the biochemical and biophysical characteristics of the different components of the plasminogen activation system. This complex system includes: the proenzymes of tissue type PA (tPA) and urokinase type PA (uPA); the active enzymes tPA, uPA and plasmin; the substrate plasminogen; several natural inhibitors of PA and plasmin activity; and the cellular receptors that bind the proenzymes, enzymes, and inhibitor-enzyme complexes. Through the coordinated interactions of these components, the location, timing, and extent of potent proteolytic activity is controlled. Recent findings on the structure, properties, biological functions, and regulation of the different components of the plasminogen activation cascade are reviewed. Current methods for assay of the amount and activity of the enzymes, inhibitors, and receptors are described. Observations implying specific functions of the system in health and disease, and its potential utilization for diagnosis are examined. Specifically, the potential application of PAs as laboratory markers of neoplasia, as diagnostic tools in diseases of the blood clotting system, their use for monitoring of thrombolytic therapy, and their possible relevance in certain disease states are described.

KEY WORDS: diabetes mellitus; extracellular matrix; fibrin; fibrinogen; fibrinolysis; fibrinolytic agents; neoplasia; plasmin; plasminogen; plasminogen activators; plasminogen activator, inhibitor; plasminogen activator, tissue type; plasminogen activator, urokinase type; receptors; rheumatoid arthritis; thrombolysis; urokinase.

Introduction t lasminogen activators (PAs) are specific serine Fminogen proteases that convert the inactive protein plasto plasmin. Through the generation of the nonspecific and potent protease plasmin, PAs affect numerous important biological processes, including:

Correspondence: Michael Mayer, Department of Clinical Biochemistry, Hadassah Medical Center and Hadassah-Hebrew Medical School, P.O. Box 12000, Jerusalem, IL-91120, Israel. Manuscript received August 23, 1989; revised November 3, 1989; accepted November 9, 1989. CLINICAL BIOCHEMISTRY, VOLUME 23, J U N E 1990

fibrinolysis and thrombolysis, cell migration, invasiveness, and metastasis. This review describes the plasminogen activation system and its relevance in h u m a n health and disease.

Components of the plasminogen activation system Figure 1 depicts the major components of the system. The PAs are produced and secreted from cells in the form of proenzymes, which are transformed to the active enzyme by a specific proteolytic cleavage. Two types of active PAs are known: the tissue type (tPA) and the urokinase-like (uPA) enzymes. Both catalyze the specific reaction, converting plasminogen to plasmin by cleavage of a single peptide bond. Both enzymes also form inactive complexes with specific inhibitors; these complexes m a y dissociate to regenerate the enzymatic activity. However, tPA differs from uPA in molecular weight, immunological specificity, and kinetic properties; the enzymes are also encoded by different genes. The details of the plasminogen activation process are reviewed in the following sections. UPA AND ITS PROENZYME

uPA has its major function in tissue-related proteolysis, and it is important in processes that entail dissolution of extracellular matrix and transversion of basement membranes (1-3). It is produced by cells and is present in the extracellular fluid in the form of an inactive single chain proenzyme (pro-uPA) (4). Conversion of pro-uPA to active two-chain uPA by catalytic amounts of plasmin is a crucial regulatory step in plasminogen activation. This conversion provides active uPA and enables an autocatalytic acceleration of uPA formation. By this mechanism, initial traces of plasmin catalyze the production of active PA, which leads to the formation of more plasmin (1-3). Active uPA is a 54 kDa protein molecule composed of two polypeptide chains of 24 kDa and 30 kDa, linked by one disulfide bond. The 24 kDa chain contains a sequence homologous to epidermal growth 197

MAYER PRO-PLASMINOGEN ACTIVATORS

PLASMINOGEN ACTIVATORS uPA

AND t P A

ACTIV[ I

~

OR(S)

ENZYME-INHIBITOR COMPt EXES

I INACTIVE} ,¥ PLASMINOGEN ACTIVATORS

( ACTIVEI

PLASMINOGEN

~) P L A S M I N

:~

=~-~-ANTIPLASMIN

FIBPlN CLOT LYSIS EXTRACELLI.ILAPPROTEOLYCJI$

Figure 1--The plasminogen activation cascade. Schematic presentation of the major steps of the cascade that lead to production ofproteolytic and/or fibrinolytic activity through activation of plasminogen. The broken line indicates conversion of the inhibited complex to active enzyme (185). Some of the reactions take place with the reactants bound to cellular receptors, to macromolecules of the extracellular matrix, or to circulating fibrin. The topography of the reactions is not shown in this scheme.

factor (EGF) and similar to a sequence present in the A chain of tPA (5). Limited proteolysis with trypsin or plasmin in vitro converts the 54 kDa high molecular weight uPA to a smaller molecular weight enzyme (31 kDa) with some loss of activity (6). uPA can be distinguished from tPA by immunological and electrophoretic methods, and by its sensitivity to the protease inhibitor aprotinin. Aprotinin inhibits both the high and low molecular weight forms of uPA in a competitive manner, but it does not inhibit tPA (7). T P A A N D I T S PROENZYME

tPA is a 70 kDa glycoprotein which is primarily active in fibrinolysis and thrombolysis (8). Its concentration in h u m a n plasma is 0.1 nmol/L. The enzyme is synthesized as a single chain molecule; its catalytic activity increases as a result of plasminmediated conversion to a two chain molecule (9). One chain is a '"heavy" 38 kDa polypeptide bearing sites responsible for enzyme regulation; the "light" 198

31 kDa chain contains the active site, similar to that of other serine proteases (10). An important feature of tPA is that its catalytic activity is markedly stimulated when the enzyme binds to certain compounds, such as, fibrin, fibrin(ogen) fragments, polylysine, heparin, or denatured proteins (5,11-14). Binding of these ligands depends on two "kringle" structures and a "finger" domain in the heavy chain (5,15,16). The activation by fibrin involves a sequential reaction of fibrin and plasminogen binding to the enzyme, which leads to formation of a cyclic ternary complex that promotes efficient clot lysis (11). The heavy chain of tPA also contains an EGF-like domain responsible for binding the enzyme to cell surface receptors (15). The domains responsible for interaction of tPA with its substrate plasminogen, with activating ligands such as fibrin, with its specific inhibitors, and with its specific receptor have been defined through the use of deletion m u t a n t s (10,17). PLASMINOGEN

Plasminogen, the physiological substrate for PAs, is a 90 kDa, single chain glycoprotein containing five homologous "kringle" structures. Plasminogen is produced in the liver (18) and in the seminiferous tubules of the testis (19); its normal plasma concentration is 2.2 ~xmol/L. The native form of plasminogen has an N-terminal glutamic acid which can easily be converted to a form with N-terminal lysine through digestion by plasmin (1,5). Plasminogen contains "lysine binding sites" (LBS) which interact specifically with certain amino acids and amino acid analogues (20). Binding of fibrin to plasminogen occurs at these sites and produces marked stimulation of the activation by either tPA or uPA (5,20,21). Plasminogen can also bind to the extracellular matrix in a specific, saturable, and reversible manner (22). Matrix bound plasminogen is a good substrate for activation by tPA or uPA. The plasmin, which is formed on, and consequently bound to, the matrix, is protected from its specific and abundant inhibitor ~2-antiplasmin (22). Therefore, binding seems to have a physiological function in the regulation of plasmin formation and activity. The plasminogen independent cleavage of certain proteins by uPA and tPA suggests that PAs m a y utilize substrates other than plasminogen. The alternative substrates include a 66 kDa matrix protein, fibronectin, and the specific inhibitor PAI-1 (23-25). Supporting a plasminogen independent function for P A s is the finding that phorbol esters induce PA activity and differentiation in HL-60 h u m a n promyelocytic leukemia cells in a system that does not require plasminogen (26). This observation suggests that other protein(s) m a y serve as natural substrate(s) for the induced enzyme. However, the physiological importance of these observations remains to be demonstrated. CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

PLASMINOGEN ACTIVATION SYSTEM

PLASMIN

Plasmin is a trypsin-like endopeptidase that hydrolyzes fibrin and other plasma proteins, as well as many extracellular matrix proteins at susceptible arginine and lysine bonds. It is a two-chain molecule, formed from single chain Glu- or Lys-plasminogen by PAs that cleave a specific Argseo-Valse 1 bond (27). The light and heavy chains of plasmin are linked by two disulfide bonds: the light chain contains the catalytic site which is similar to that of other serine proteases; the heavy chain contains a regulatory domain (20). Plasmin activity is subject to inhibition by binding to its specific inhibitor ~2-antiplasmin (28). Plasmin, like its parent molecule plasminogen, contains LBS that interact with lysine, 6-aminohexanoic acid, tranexamic acid, fibrinogen, fibrin, and related compounds (5). The LBS are located in the protein structure in an area that is distinct and distant from the catalytic site (5,29). Nonetheless, binding of ligands to LBS produces modulatory responses that affect the catalytic activity of the enzyme (30,31). Binding of ligands, such as the amino acid analogue 6-aminohexanoic acid and fibrin to the LBS, also protects plasmin against inhibition by a2-antiplasmin (32,33). This property is responsible for the relative resistance of plasmin, formed on the fibrin clot surface, to inhibition by ~2-antiplasmin. In contrast, plasmin generated in the circulating blood is rapidly neutralized by the inhibitor (8). RECEPTORS Binding of uPA to a specific receptor on the cell surface appears to be important for localization of uPA-catalyzed plasminogen activation (34,35). Binding is rapid, saturable, and with a high affinity of ~ 1 0 - lO M. The bound enzyme is not internalized or rapidly degraded (35,36). Binding of uPA to the receptor does not involve the catalytic site of uPA; it occurs at the EGF domain of uPA and pro-uPA (35). Therefore, bound uPA retains its enzymatic activity (35,36). uPA receptors have been found in monocytes and in several cultured h u m a n cell lines such as, fibroblasts, breast carcinoma, and umbilical vein endothelial cells (34-37). Affinity of binding and concentration of binding sites varies in different cells, subject to regulation by modulators such as tumor promoters, EGF, and dimethylformamide (3538). The receptor has been recently purified and shown to be a single chain, 55-60 kDa, heavily glycosylated protein (37). Single chain pro-uPA binding to the cellular receptors is followed by conversion of the bound enzyme to active, two chain uPA on the surface of the cells (39). Receptor bound uPA can still bind PAI-1 and maintains its susceptibility to inhibition by this inhibitor (40). The surface receptor also binds PA-inhibitor complexes such as uPA-PAI-1 (40).

CLINICALBIOCHEMISTRY,VOLUME23, JUNE 1990

In addition to having receptor sites that bind the enzyme, various cells are also equipped with receptors on the cell surface which bind the substrate, plasminogen (41,42). Binding ofplasminogen is characterized by its low affinity, and seems, at least in part, to involve sites which also bind plasmin (43). Cell bound plasminogen undergoes activation to plasmin by receptor bound uPA, while catalytic amounts of plasmin at these sites serve to produce active uPA from bound pro-uPA. This mechanism is responsible for an autocatalytic process that is subject to regulation by specific inhibitors such as PAL1 and PAI-2 (see below). Cell bound plasmin is refractory to inhibition by its specific inhibiter ~2-antiplasmin (44). Thus, plasminogen activation on receptor sites results in the cells ability to promote pericellular proteolysis. The newly produced proteolytic activity is localized because plasmin generation is limited to the close environment of the cell surface (3,35). Receptors for tPA also exist, and binding of tPA to its specific cellular receptors does not cause loss of the enzymatic activity (45). Simultaneous binding of pro-uPA, uPA, plasminogen and plasmin at the cell surface produces a topographical proximity which is important for the biological function of the plasminogen activation process (3,42,43). The proximity enables efficient autocatalytic activation of plasminogen by uPA, and serves to localize the proteolytic activity that emerges via plasmin formation (1,3,35). Thereby, the cells are armed with a matrix-degrading activity, which is an essential tool for cell migration into solid tissues. Because the receptor bound uPA is distinctly localized at cell-cell and cell-substratum contacts (46,47), the receptors control the position of plasmin activity at focal contact sites. Cell invasiveness and cell migration depend on this focal localization of the components (35). INHIBITORS Emergence of premature and uncontrolled activity of PAs, and activity at inappropriate site(s), is potentially damaging. Time- and site-specific inhibition of the catalytic activity of PAs is, therefore, instrumental in the prevention of aberrant plasminogen activation. Similarly, activity of the newly generated plasmin has to be strictly regulated to prevent uncontrolled proteolysis. It is, therefore, not surprising that potent and specific inhibitors of PAs and plasmin are present in cells, in the circulation, and in other extracellular compartments. The major function of these inhibitors is to regulate the catalytic activities since the balance between enzymes and inhibitors determines net activity. In the circulation, PA inhibitors block premature fibrinolysis (48-50). Plasma levels of the plasminogen activator inhibitors type 1 and type 2 (PAL1 and PAI-2, see below) are elevated in conditions associated with enhanced tendency for thrombus formation. Since

199

MAYER

plasminogen activation is also important in tissue remodelling, neoplasia and invasiveness (see below), the inhibitors may also be implicated in cancer. Several recent studies have demonstrated the ability of natural and synthetic inhibitors of PAs to suppress tumor cell invasion in in vitro experimental models of metastasis (51-53). Various factors might complicate the interaction between inhibitors and the enzymes; therefore, a direct correlation with enzyme/inhibitor ratios and activities is not always observed. For example, binding of tPA to fibrin reduces the ability of PAL1 and PAI-2 to react with the enzyme. In addition, various compounds that affect the PA system do so by independent effects on the enzymes and inhibitors; therefore the final result, in terms of net activity, is not easily predictable (54). In the following section a brief review on specific and potent inhibitors of PAs and plasmin is presented. For more information, refer to recent reviews (48,49). PAI-1

PAI-1 is a 50 kDa single-chain plasma glycoprotein that rapidly and specifically forms equimolar, inactive complexes with one- or two-chain tPA, two-chain uPA, and the anticoagulant protein C, but not with single chain uPA or with plasmin (55,56). PAI-1 has a high second order rate constant for tPA or uPA, and Kd values of 8 × 10-13 M for tPA and 2 × 10- is M for uPA. Therefore, PAI-1 appears to be the physiological inhibitor of PAs. PAL1 has significant homology with members of the serine protease inhibitor (Serpin) family (57). Serpins share structural features, a common functional principal, and are important regulators of the processes of coagulation, inflammation, and complement activation (58). The 1:1 complex formed between PAI-1 and tPA or uPA resists dissociation by denaturing agents such as urea or sodium dodecyl sulphate; however, these agents regenerate activity of the bound enzyme (59). Although PAI-1 is synthesized in an active form, it is rapidly converted to an inactive (latent) variant (60). This inactivation can be partially reversed by sodium dodecyl sulphate, guanidine, and urea (59,60). The physiological relevance of the active and latent forms of the inhibitor remain to be determined. PAI-1 is present in platelets, placenta, serum, and is produced by endothelial cells, monocytes/macrophages, and various normal and neoplastic cells in culture (61,62). Vascular and liver endothelial cells and platelets seem to be the major sources of the inhibitor in the body (50,59,63). The gene for PAI-1 is located on chromosome No. 7 (64). Production of the inhibitor is modulated by a variety of hormones and growth factors (see Table 2). The elevations in PAI-1 protein level as a result of exposure to cytokines, growth factors, and hormones are often preceded by elevations in PAI-1 mRNA level; this

200

indicates transcriptional regulation (54,65,66). PAI-1 activity (and antigen) level in the blood shows an extremely large range. Activities between 0.0-1.3 nmol/L have been found in the plasma of healthy subjects (50,67,68). Activity of PAI-1 in plasma correlates with plasma insulin levels (69) and increases during pregnancy (70). PAI-1 plays an important role in the development of arterial and venous thromboembolism, and it also acts as an acute phase reactant (see below). In plasma, PAI-1 is bound to a protein that was recently identified as vitronectin (71). It also binds to vitronectin in the extracellular cell matrix (72), and is deposited by cultured endothelial cells into the substratum matrix. A large part of PAI-1 in the substratum is associated with the adhesive proteins vitronectin and fibronectin (72,73). PAI-2

PAI-2 is a 46,000 kDa serine protease inhibitor with a higher affinity for uPA than for tPA. The cDNA encoding for PAI-2 in human monocytic cell line U937 has been characterized (74). The gene for PAI-2 is located on chromosome 18q21-23, has two common alleles, and is highly homologous to chicken ovalbumin gene (75). PAI-2 is hardly detectable in nonpregnant plasma, while levels of up to 100 ~g/L (2 ~mol/L) are observed in the third trimester of pregnancy (50,76). Because PAI-2 is found mainly in placental tissue, this suggests a function for the inhibitor in regulation of hemostasis during pregnancy and parturition (77). High levels of the inhibitor may produce microthrombi in the placenta that lead to impaired fetal blood supply and consequently, to retarded intrauterine growth (77). PAI-2 is induced by tumor necrosis factor (TNF), phorbol esters and endotoxin; this suggests its involvement in local inflammatory processes (78,79). Based upon kinetic considerations, PAI-2 is probably more relevant as an inhibitor of uPA than of tPA. It is a serpin that does not inhibit pro-uPA or plasmin. In many cell types, PAI-2 is located intracellulary. The latter observation raises a question concerning the physiological function of PAI-2, since its natural substrates uPA and tPA are mainly extracellular components. Protease nexin

Produced by fibroblasts, heart muscle cells, and kidney epithelial cells, protease nexin is a 51 kDa glycoprotein that inhibits serine proteases such as trypsin, thrombin, plasmin, uPA, single and twochain tPA, but not pro-uPA (80,81). The absence of protease nexin in circulating blood and its relatively unfavorable affinities toward PAs suggest that it does not fulfill a major function in controlling plasminogen activation. The main function of protease nexin appears to be facilitation of endocytosis of proteases. This assumption is based on observations

CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

PLASMINOGEN ACTIVATION SYSTEM

that protease nexin forms covalent complexes with several proteases including uPA and mediates their uptake by cells (48,49).

c~2-antiplasmin cx2-antiplasmin is a 67 kDa single-chain glycoprotein with a plasma concentration of 1 ~Lmol/L. It is an extremely active inhibitor, with a high second order rate constant. By binding to the light chain of plasmin, a 1:1 stoichiometric inactive complex is formed. Ligands such as fibrinogen, lysine, and 6-aminohexanoic acid, prevent interaction between the inhibitor and the enzyme (5,8,32). The inhibitor is synthesized and secreted by liver cells (82); it is the most abundant and active natural inhibitor of plasmin activity. The other plasma proteinase inhibitors do not seem to play a role in the inactivation of blood plasmin (20).

Assays of the components ENZYMES

Plasminogen activation can be measured either by direct assays which follow the action of PAs upon specific substrates, or by coupled, indirect methods. A widely used direct method measures the amidolytic activity of PAs by following the cleavage of chromogenic substrates, such as pyro-glu-gly-argp-nitroanilide, to produce the chromophore p-nitroanilide (83). Another direct assay measures the ability of PAs to convert the single chain of plasminogen to the two peptide chains of plasmin. By reduction of a single disulfide bond, the heavy and light chains can be separated on SDS-PAGE. When the plasminogen is labeled with ~25I, its conversion to plasmin can be quantitated (84). A common disadvantage of the direct assays is that nonspecific proteases might also contribute to the measured activity. Indirect assays of PAs involve a dual-stage reaction. Plasminogen is first converted to plasmin by the assayed enzyme, and the plasmin activity formed is subsequently determined. The advantage of the indirect assays is that only plasminogen-dependent activity is measured, and therefore only net plasminogen activation is determined. A widely used method is the fibrin plate assay that follows generation of plasmin-mediated fibrinolytic activity (85). In this method, PA activity solubilizes fibrin through production of plasmin. Area of lysis on layers of fibrin/ agar plates is measured as an estimate of plasmin activity in a system containing plasminogen. The fibrin plate assay is highly specific and easily quantitated. It utilizes the physiological substrate fibrin and thereby mimicks the fibrinolytic process that occurs in the body (85). When 125I-labeled fibrin is used, fibrinolysis can also be measured quantitatively, with an even higher sensitivity. The disad-

CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

vantage of the fibrinolytic methods is that they are time consuming; the fibrin frequently contains impurities such as proteases and thrombin which contribute to hydrolysis; also, the formed plasmin tends to absorb to fibrin, causing an underestimation of the plasmin level. Insolubility of the fibrin substrate is an additional complication because it results in nonlinear kinetics. Alternative coupled assays follow plasmin formation by its ability to degrade other proteins, such as casein (86) or globin (14). The latter method was useful for studies in different experimental systems (14,26,87-89). Numerous chromogenic substrates are available for the determination of PAs by the plasmin-mediated reaction. Substrate cleavage is followed in a parabolic rate assay (90), or in an end point assay with or without fibrin as a stimulator (91,92). Some of these methods enable assays in plasma without prior precipitation of the euglobulin fraction, while others use the euglobulin fraction (93). Sensitive immunological methods have been devised which utilize specific antibodies for the detection oftPA or uPA antigens, and ELISA methods for use in plasma and other biological fluids (94-96). These reactions provide unmatched specificity and sensitivity. RECEPTOR

The affinity and number of receptors are usually determined by using labeled ligands such as 125Iurokinase, or labeled fragments of the ligands containing the binding site sequence, such a s 125Iamino terminal fragment of urokinase (34,35,37,40). INHIBITORS

In view of the growing interest in the higher risk for thrombosis in individuals displaying increased PAL1 in plasma (see below), reliable analytical methods to measure plasma inhibitors are desired. The inhibitors are often assayed by monitoring their ability to reduce the enzyme activity, measured by the conventional assays of uPA or tPA as described above. For that purpose, standard PA assays are performed in the absence or presence of the inhibitor(s). From the residual activity remaining in the presence of the inhibitor, inhibitory activity can be deduced (92). A simple standard assay for plasma PAI-1 activity follows the inhibition of one chain tPA by serial dilutions of added plasma (97). This assay provides good analytical recovery, has a low detection limit, and is precise. A unique "reversed zymography" system traces PA-inhibitory activity on acrylamide gel electrophoresis. In this assay, lysis of fibrin/agar films is prevented and a lysis-resistant zone appears in regions of the gel where the inhibitor is present. The tested proteins are electrophoresed in polyacrylamide gels containing sodium dodecyl sulphate. The

201

MAYER

gels are subsequently covered with a fibrin overlay containing plasminogen and uPA. The inhibitor(s) appear as zone(s) which inhibit the PA-induced lysis of the fibrin overlay. This technique also provides an estimation of the apparent molecular weight of the inhibitory proteins (98). The inhibitory proteins can also be determined using specific monoclonal and polyclonal antibodies (62,99).

Biological functions FIBRINOLYSIS

The major physiological function of plasminogen activators in the circulation is to provide plasmin that will dissolve blood clots. The ability of PAs to produce plasmin is also the basis for their increasing application in medicine. The use of PAs to treat patients with acute myocardial infarction is based on clinical evidence that occlusive coronary thrombus precipitates most infarctions. Recanalization of the occluded blood vessels can often be achieved by the early administration of the fibrinolytic enzymes. Large clinical trials show improved survival rates after myocardial infarction in patients receiving different types of thrombolytic enzymes, including tPA or uPA (100-104). Thrombolytic therapy with different PAs is also successfully applied in the treatment of deep vein thrombosis, pulmonary embolism, peripheral arterial occlusion and several additional conditions associated with thrombosis (103). The fibrin affinity of the thrombolytic enzyme tPA is an important pharmacological attribute since binding of tPA to plasminogen in the presence of fibrin not only contributes to localization of the catalytic activity to the clot but also increases catalytic efficiency. Thus, fibrin selectivity is important for effective fibrin clot localized thrombolysis without marked activation of systemic plasminogen and without induction of a general hemostatic defect. Due to its fibrin selectivity, tPA appears to have an apparent advantage over uPA or streptokinase in clinical use with cases of localized thrombotic occlusions. It was recently suggested that in physiological fibrinolysis, tPA and pro-urokinase may act in a sequential, complementary, and synergistic manner to maintain the fibrin specifity of plasminogen activation. Such synergistic fibrinolytic effects can potentially be applied as thrombolytic combination therapy with these agents (105). A detailed discussion of the function of plasminogen activation in fibrinolysis, and its importance in clinical and therapeutic aspects of thrombolysis is beyond the scope of this review. This subject has been extensively reviewed (5,8,20,33,103-107). DEGRADATION OF THE EXTRACELLULARMATRIX

uPA plays a major role in tissue destruction and cell migration because it promotes extracellular 202

matrix degradation (1-3,108). This effect is catalyzed by a local, uPA-mediated production of plasmin and probably by additional proteolytic enzymes that are activated by plasmin and/or PAs. Emergence of matrix degrading activity was studied in a variety of tissue and cellular processes, and the invasion of the matrix by neoplastic cells was shown to involve protease induced degradation of the cell substratum (108-110). In a model system of invasion, a requirement for a proteolytic cascade has been observed (53,109). Experimental support for the role of PAs in matrix destruction as a functional stage in invasion also comes from other observations: transformation by oncogenic viruses induces uPA production; uPA activity is involved in tissue involution in many malignant and nonmalignant conditions; and PAs are characteristically localized in areas of tumor invasion (1-3,110). Further support for this is that anticatalytic antibodies against uPA reduce invasiveness and incidence of metastasis (51-53,111), and inhibit the growth pattern and morphological changes that characterize virus induced transformation (112). The finding that plasmin converts procollagenase to active collagenase indicates that in addition to being a direct mediator of matrix cleavage, uPA also recruits additional proteolytic enzymes that can cleave specific matrix proteins (51-53,108,113). Similarly, PA and heparanase were shown to participate in the sequential degradation of the extracellular matrix (114). The distinct pericellular localization of the different components of the plasminogen activation system also supports the role of PAs in cell migration, uPA is found at focal cell-cell and cell-substratum contact sites (46,47,72); whereas, PAI-1 is distributed homogeneously in the matrix under the cells (46), and is present in the growth substratum even before it appears in the culture medium of cells (115). Since cell migration depends on the sequential formation and dissolution of cell-substratum contacts, the specific localization of PAs and plasmin assists in the control of the migration process. The plasmin formed is expected to promote the hydrolysis of bonds between matrix proteins and cells, and to mediate cleavage of adhesive contact proteins. The matrix proteins which appear to be involved in these processes are vitronectin, fibronectin and laminin. In this context, it is interesting that PAI-1 colocalizes with vitronectin in the matrix (46), and that PAI-1 binds to vitronectin (72). Thus, focalized distribution of PAs and their inhibitors in the extracellular space and their specific interaction with matrix components confines and localizes extracellular proteolysis and is .most probably involved in cell movements (3). Net plasminogen activation is a result of an intricate balance between receptors, proenzymes, enzymes, and inhibiters. Although each of these components can be independently and differentially regulated in time and space, together they can direct the activity to specific site(s) and to specific timing, according to requirement. Table 1 lists some important biological CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

PLASMINOGEN ACTIVATION SYSTEM

TABLE 1

Studies Implicating PAs in Biological Processes Other Than Fibrinolysis Organ/Process

Suggested Function

Mammary gland Implantation

Postlactation involution (152) Trophoblast invasion following implantation of fertilized ovum (153) Differentiation, wound re-epithelialization, tissue destruction in psoriasis (154) Gestation (155) Ovulation, oocytematuration (156,157,158) Spermatogenesis (159,160) Angiogenesis (161) Tumor invasion and metastasis (1,2,53,108) Migration, inflammation (1,61,162) Involution post castration (163) Prevention of adhesion (88)

Keratinocytes Amniotic fluid

Ovaries Testes Capillaries Neoplasia Macrophages Prostate Peritoneum

systems which involve plasminogen activation. For more information on these observations see refs. 1-3,110 and the specific citations in Table 1.

Regulation Plasminogen activation is under stringent metabolic control; several sites and mechanisms of regulation have been observed. Among the regulating factors are the specific, high affinity, and fast acting inhibitors PAI-1 and PAI-2. These inhibitors are by themselves subject to regulation at the levels of gene expression, activity, and secretion (33,48,49). Binding to receptors on the cell surface is an additional means of regulation, limiting the activity to specific sites and modifying the mutual interactions of the components and their response to other modulators. Another regulatory mechanism is related to the existence of latent enzyme forms, which are either inactive proenzymes or enzyme-inhibitor complexes. For example, the suppressive effect of dexamethasone on PA activity in cultured cardiac myocytes involves formation of an inactive complex and/or production of an inactive proenzyme (87). Inactive enzyme forms can, under certain conditions, such as exposure to denaturants or proteolytic cleavage, be converted to their active counterparts (1,2,87,88, 106,107,116). The hormones, cytokines, and growth factors listed in Table 2 regulate plasminogen activation by modulating the level of synthesis and/or activity of the components of the system. In addition to regulation by gene expression and through covalent or noncovalent binding to inhibitors and activators, various ligands affect PAs and plasmin activity by inducing conformational changes in the enzymes and/or substrates (20,21,29-32, 106,107,117,118). These conformational changes markedly affect the catalytic activity of the activators and ofplasmin. For example, tPA contains LBS, a flbrin(ogen) binding site and a site that binds with LBS (118). Binding of ligands to these sites allosterically affects the catalytic domain of the enzyme (117,118). CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

Likewise, plasmin undergoes regulation at a site different from the catalytic site by interacting with ~-lactam antibiotics (119). The kinetics of activation of single chain uPA by plasmin, similarly indicate negative cooperativity (120). Susceptibility of plasmin to ~2-antiplasmin is also conformationally controlled by the availability of LBS on the enzyme (32,121). These conformational changes therefore render the enzyme subject to regulatory modulation.

Relevance in clinical biochemistry M A R K E R OF NEOPLASIA

Numerous studies examined the occurence of PAs in neoplasia, their relation to metastasis formation, transformation, and their localization in particular tumors and cell types (1,2,108). Due to the potential role ofproteolytic degradation in neoplasia, researchers sought clear correlations between cell malignancy or spreading, and cell PA activity. Indeed, in certain tumor cell lines, PA activity correlates with the metastatic potential, and PA appears to be an index of the ability of the tumor cells to colonize target organs (122,123). However, these associations remain to be confirmed. A problem in establishing a causal relationship between neoplasia and PA activity is that normal growth processes also involve plasminogen activation (2). Furthermore, conflicting results were obtained in studies on potential correlations between PA activity of tumor cell lines in vitro and their metastatic capacity in vivo (2). Production of PA is probably neither necessary nor sufficient for tumor formation, although plasminogen activation is somehow involved in invasion and metastasis. Recent work supports the involvement of uPA in cancer by demonstrating that antibodies, which inhibit the catalytic activity ofuPA, abort the invasiveness of tumor cells and prevent metastasis (51,53). According to Dano et al. (2), it appears that consistent, but still inconclusive, evidence supports the assumption that uPA production in cancer cells 203

MAYER TABLE 2 Regulation of the PA System by Hormones, Growth Factors, and Cytokines

Component

uPA

Regulator 3',5'-Cyclic adenosine monophosphate (cAMP) (164)

Retinoic acid (165) Phorbol ester (164) Glucocorticoids (148,166,167) Estrogens (168) Tumour necrosis facter-~ (TNF-cx), interleukin-l, lymphotexin (169) tPA

Glucocorticoids (170) Follicular stimulating hormone (FSH), gonadotropinreleasing hormone (GnRH) (157) Phorbol ester (171) cAMP (170)

Receptor

Phorbol ester (34,36,37) Formamide (38) Epidermal growth factor (EGF) (34,36)

PAI-1

Phorbol ester (54,62) Glucocorticoids (62,65,172,173) TNF-~ (174) Thrombin (175) Transforming growth facter-~ (TGF-~) (176) Endotoxin (177,178) Interleukin (179,180,181) Luteinizing hormone (LH) and FSH (157) EGF (182)

PAI-2

TNF, phorbol esters, endotoxin (183) Endotoxin (78,79) Phorbol ester (184) Glucocorticoids (65)

mediates proteolysis of extracellular matrix. However, use of PAs as a clinical marker for neoplasia, invasive growth or metastasis is presently inappropriate. Still, under specific circumstances, the PA system might serve a diagnostic function in malignant diseases. For example, the production of tPA and uPA by leukemic cells can be used to predict prognosis and response to chemotherapy in patients with acute myeloid leukemia (124). Patients whose cells produce only tPA have a lower survival and fail to respond to chemotherapy; uPA producers have a longer survival and a better response to treatment. The production of tPA seems to characterize a more primitive cell, while development of a more specialized, differentiated cell is associated with uPA production. On the basis of this finding, the type of PA released by peripheral blood cells may indicate the leukemic state (125). DIAGNOSIS OF DEFECTS IN CLOT LYSIS

Since excessive fibrinolysis causes bleeding while reduced fibrinolysis causes thrombosis, it is not 204

surprising that maintenance of a proper balance between components of the PA system is crucial for normal clot formation and dissolution. Defciencies in plasma plasminogen, low release of tPA from endothelial cells, abnormal fibrinogen in plasma, increased circulatory levels of PAI-1 and PAI-2, and deficiency of circulating a2-antiplasmin are some of the biochemical abberations of fibrinolysis that have been described. In general, thrombotic patients often exhibit low fibrinolytic activity due to deficient tPA or to elevated PA-inhibitor (126-129). Some defects in clot lysis are directly, and most probably specifically, related to the changes in PA inhibitor activity. For example, in deep vein thrombosis, higher than normal levels of PA inhibitor are found (130-137). An elevated blood PAI-1 level is clearly an independent risk factor for impaired fibrinolytic function in deep vein thrombosis, disseminated intravascular coagulopathy and myocardial infarction (67,130,133,137). By contrast, PAI-1 level is generally decreased in bleeding diatheses. PAI-1 as a risk factor in myocardial infarction is supported by observations that myocardial infarction is associated CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

PLASMINOGEN

ACTIVATION SYSTEM

with increased PA-inhibitory activity and decreased levels of tPA (67,132,133,137), and by high PAI-1 levels found in young survivors of myocardial infarction (138). These observations clearly suggest that the balance between circulatory tPA and PAI-1 may regulate vascular thrombolysis, and that high levels of plasma inhibitors correlate with higher probability of thromboembolism. It is anticipated that the PA-inhibitor level in plasma will be used as a useful marker to evaluate the risk of developing deep vein thrombosis (135). While the relation between PAI-1 and thrombotic diseases is likely to reflect an effect of the inhibitor on clot lysis, other studies suggest that PA-inhibitory activity is nonspecifically increased in a number of disease states. These studies imply that a raised level of the inhibitor is not associated with any specific disease, and may reflect a general response to acute illness (49). This is supported by the increased PAI-1 antigen and PAI-1 activity found in patients with malignant tumors, hepatic insufficiency, after extracorporeal circulation in cardiac bypass surgery, after major surgery, and in severe trauma and septicemia (50,67, 131-133). Laboratory assays of components of the plasminogen activation system in plasma should elucidate the molecular basis of fibrinolytic defects and assist in detecting patients at risk of thromboembolism. It is hoped that further research will indicate therapeutic means to minimize undesired changes in fibrinolytic activity that mediate thrombosis. The potential application of orally-administered stanazolol for enhancement of fibrinolytic activity by its stimulatory effect on endogenous tPA activity is a step toward this goal (139). MONITORING THERAPY WITH THROMBOLYTIC AGENTS

Numerous studies have led to current interest in the clinical use of plasminogen activating agents in thromboembolic conditions, pulmonary embolism, deep vein thrombosis and myocardial infarction (5,103,104). At least five types of active fibrinolytic enzymes are currently being evaluated or used for thrombolytic treatment of acute myocardial infarction and pulmonary embolism. These include: streptokinase-plasminogen, uPA, tPA, single chain uPA, and acylated plasminogen-streptokinase complex. The relative advantages and disadvantages in the therapeutic use of each of these agents are under study (5,140). The increased use of any of these agents for clinical therapy will involve closely monitoring the circulatory level of the components of the plasminogen activation system. Such monitoring is required, since excessive thrombolytic activity is likely to produce the untoward side effects of bleeding, and particularly, cerebral hemorrhage. Routine laboratory tests for clinical follow up have yet to be defined and properly established. Monitoring of the coagulation status and extent of flbrinolysis as routine laboratory procedures have been recommended CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

during therapy with thrombolytic agents (103,141). Direct determination of plasminogen activation activity in blood samples of patients undergoing fibrinolytic therapy will be an important part of patient monitoring in the future. DIABETES MELLITUS

Vascular complications and disorders of hemostasis represent the most important causes of morbidity and mortality in diabetic patients (142-144). Increased plasma insulin predisposes to atherosclerosis (143). Reduced blood fibrinolytic activity has been identified as a factor that contributes to atherosclerosis (138); variation in plasma insulin concentration, even in the physiological range, modulates the fibrinolytic system at the PA inhibitor level (69). Therefore, the effect of insulin on atherosclerosis could be mediated through its effect on PA inhibitor level. In diabetes mellitus, the normal balance between coagulative and fibrinolytic activity is affected, but no major abnormalities have been reported in levels and activities of the PA system in the plasma of diabetic patients (145). By contrast, abnormally low fibrinolytic activity is produced by vessels from diabetics, and a reduced production of PA in response to venous occlusion occurs in diabetic patients (144). ~2-antiplasmin activity and antigen levels in diabetic patients fall within normal ranges, indicating that this inhibitor is unaffected in this disease (146). Fibrin enhanced plasmin formation in an in vitro plasminogen activation assay was markedly impaired when the plasminogen or the tPA was obtained from patients with uncontrolled diabetes (145); however, the deranged function improved upon normalization of the blood glucose level (147). Therefore, the fibrinolytic defects are secondary to the metabolic consequences of diabetes and are not directly related to the disease (147). Contrasting reports on the implication of PAs in diabetes currently preclude use of these assays in the clinical evaluation of fibrinolysis in diabetic patients. RHEUMATOID ARTHRITIS

Several findings suggest a link between the PA and plasmin activities and the pathogenesis of rheumatoid arthritis (148-150). The PA system is known to be involved in inflammatory processes of different etiologies, and has been found in rheumatoid synovial fluid and tissue. Plasmin can degrade cartilage and activate latent collagenase; anti-inflammatory glucocorticoids that are clinically effective in the treatment of rheumatoid arthritis suppress uPA activity in synovial fibroblasts (148); elevated plasmin levels are found in rheumatoid synovial fluid (149). Cartilage destruction in rheumatoid arthritis involves production of proteases by synovial cells. On the basis of these observations, PA appears to assist in cartilage destruction associated with rheumatoid arthritis. The finding that synovial cell PA 205

MAYER production is stimulated by interleukin-1 (150) furt h e r supports its role as an i m m u n e mechanism in the induction of synovial cell PA. These observations have not yet been applied for diagnostic or therapeutic purposes. Conclusions

The subject of plasminogen activation offers a fascinating area of research because it combines basic science with medically r e l e v a n t and clinical laboratory oriented research. F u t u r e studies in this field should clarify some of the issues discussed. F u t u r e work is likely to examine intriguing new possibilities, such as the apparent association between thrombosis, coronary atherosclerosis, and hyperlipoproteinemia (151). Application of knowledge gained in the area of plasminogen activation will be used for diagnostic and therapeutic purposes; these include: use of components of the system for the diagnosis of cancer, early detection of p r i m a r y tumors and metastasis, and t r e a t m e n t of neoplastic diseases. Similarly, the role of plasminogen activation in cardiovascular and thrombotic conditions will be clarified and applied to diagnosis and therapy.

References

1. Saksela O. Plasminogen activation and regulation of pericellular proteolysis. Biochim Biophys Acta 1985; 823: 35-65. 2. Dano K, Nielsen LS, Pyke C, Kellerman GM. Plasminogen activators and neoplasia. In: Kluft C, ed. Tissue-type plasminogen activator (tPA): physiological and clinical aspects. Pp. 19--46. Boca Raton: CRC Press, 1988. 3. Dano K, Behrendt N, Lurid LR, et al. Cell surface plasminogen activation. In: Schirrmacher V, SchwartzAlbiez R, eds. Cancer metastasis: molecular and cellular biology, host immune responses and perspectives for treatment. Pp. 98-107. Berlin: SpringerVerlag, 1989. 4. Petersen LC, Lund LR, Nielsen LS, Dano K, Skriver L. One-chain urokinase-type plasminogen activator from human sarcoma cells is a proenzyme with little or no intrinsic activity. J Biol Chem 1988; 263: 11189-95. 5. Verstraete M, Collen D. Thrombolytic therapy in the eighties. Blood 1986; 67: 1529-41. 6. Barlow GH, Francis CW, Marder VJ. On the conversion of high molecular weight urokinase to the low molecular weight form by plasmin. Thromb Res 1981; 23: 541-7. 7. Lottenberg R, Sjak-Shie N, Fazleabas T, Roberts RM. Aprotinin inhibits urokinase but not tissue-type plasminogen activator. Thromb Res 1988; 49: 549-56. 8. Collen D. Molecular mechanisms of fibrinolysis and their application to fibrin-specific thrombolytic therapy. J Cell Biochem 1987; 33: 77-86.

206

9. Ranby M, Bergsdorf N, Nilsson T. Enzymatic properties of the one- and two-chain form of tissue plasminogen activator. Thromb Res 1982; 27: 175-83. 10. Ny T, Elgh F, Lund B. The structure of the human tisaue-type plasminogen activator gene: correlation of intron and exon structures to functional and structural domains. Proc. Natl Acad Sci USA 1984; 81: 5355-9. 11. Hoylaerts M, Rijken DC, Lijnen HR, Collen D. Kinetics of the activation of plasminogen by human tissue plasminogen activator. Role of fibrin. J Biol Chem 1982; 257: 2912-9. 12. Nieuwenhuizen W, Vermond A, Voskuilen M, Traas DW, Verheijen JM. Identification of a site in fibrin(ogen) which is involved in the acceleration of plasminogen activation by tissue-type plasminogen activator. Biochim Biophys Acta 1983; 748: 86-92. 13. Allen RA. Enhancing effect of poly-lysine on the activation of plasminogen. Thromb Haemost 1982; 47: 41-5. 14. Finci Z, Nachshon I, Sharoni Y, Mayer M. Functional assay of plasminogen activator by hydrolysis of laC-globin. Exp Hematol 1986; 14: 293-7. 15. Rijken DC, Groeneveld E. Isolation and functional characterization of the heavy and light chains of human tissue type plasminogen activator. J Biol Chem 1986; 261: 3098-102. 16. Ichinose A, Takio K, Fujikawa K. Localization of the binding site of tissue-type plasminogen activator to fibrin. J Clin Invest 1986; 78: 163-9. 17. Van Zonneveld A-J, Veerman H, MacDonald ME, Van Mourik JA, Pannekoek H. Structure and function of human tissue-type plasminogen activator (tPA). J Cell Biochem 1986; 32: 169-78. 18. Raum D, Levey R, Taylor PO, Starzl TE. Synthesis of human plasminogen by the liver. Science 1980; 208: 1036-7. 19. Saksela O, and Vihko KK. Local plasminogen synthesis by the seminiferous tubules of the testis. F E B S Lett 1986; 204: 193-7. 20. Collen D. On the regulation and control of fibrinolysis. Thromb Haemost 1979; 41: 77-89. 21. Takada A, Sugawara Y, Takada Y. Enhancement of the activation of glu-plasminogen by urokinase in the simultaneous presence of tranexamic acid and fibrin. Haemostasis 1989; 1: 26-31. 22. Knudsen B, Silverstein RL, Leung LLK, Harpel PC, Nachman RL. Binding of plasminogen to extracellular matrix. J Biol Chem 1986; 261: 10765-71. 23. Keski-Oja J, Vaheri A. The cellular target for the plasminogen activator, urokinase, in human fibroblasts. 66,000 dalton protein. Biochim Biophys Acta 1982; 720: 141-6. 24. Quigley JP, Gold LI, Schwimmer R, Sullivan LM. Limited cleavage of cellular fibronectin by plasminogen activator purified from transformed cells. Proc Nat! Acad Sci USA 1987; 84: 2776-80. 25. Nielsen LS, Andreasen PA, Grondahl-Hansen J, Skriver L, Dano K. Plasminogen activators catalyze conversion ofinhibitor from fibrosarcoma cells to an inactive form with a low apparent molecular weight mass. F E B S Lett 1986; 196: 269-73. 26. Kidron M, Nachson I, Mayer M, Fibach E. Plasminogen activator activity in differentiating leukemia cells. F E B S Lett 1984; 177: 66-70. 27. Robbins KC, Summaria L, Hsieh B, Shah RJ. The

CLINICAL BIOCHEMISTRY,VOLUME 23, JUNE 1990

PLASMINOGEN ACTIVATION SYSTEM

28. 29. 30. 31. 32.

33. 34. 35. 36.

37.

38.

39.

40.

41. 42.

43. 44.

45. 46.

peptide chains of human plasmin. Mechanism of activation of human plasminogen to plasmin. J Biol Chem 1967; 242: 2333-42. Wiman B, Collen D. On the kinetics of the reaction between human antiplasmin and plasmin. Eur J Biochem 1978; 84: 573-8. Rickli EE, Otavsky WI. A new method of isolation and some properties of the heavy chain of human plasmin. Eur J Biochem 1975; 59: 441-7. Higazi A, Mayer M. Effect of penicillins and 6aminohexanoic on the kinetics of human plasmin. Biochem J 1989; 260: 609-12. Higazi A, Mayer M. Inhibition of plasmin by fibrinogen. Biochem J 1990; in press. Wiman B, Lijnen HR, Collen D. On the specific interaction between the lysine-binding sites in plasmin and complementary sites in ~2-antiplasmin and in fibrinogen. Biochim Biophys Acta 1979; 579: 14254. Ranby M, Brandstrom A. Biological control of tissue plasminogen activator-mediated fibrinolysis. Enzyme 1988; 40: 130-43. Blasi F, Stoppelli P, Cubellis MV. The receptor for urokinase-plasminogen activator. J Cell Biochem 1986; 32: 179-86. Blasi F. Surface receptors for urokinase plasminogen activator. Fibrinolysis 1988; 2: 73-84. Estreicher A, Wohlwend A, Belin D, Schleuning W-D, Vassali J-D. Characterization of the cellular binding site for the urokinase-type plasminogen activator. J Biol Chem 1989; 264: 1180-9. Nielsen LS, Kellerman GM, Behrendt N, Picone R, Dano K, Blasi F. A 55000-60000 M r receptor protein for urokinase-type plasminogen activator. Identification in human tumor cell lines and partial purification. J Biol Chem 1988; 263: 2358-63. Boyd D, Florent G, Murano G, Brattain, M. Modulation of the urokinase receptor in human colon cell lines by N,N-dimethylformamide. Biochim Biophys Acta 1988; 947: 96-100. Cubellis MV, Nolli ML, Cassani G, Blasi F. Binding of single-chain prourokinase to the urokinase receptor of h u m a n U937 cells. J Biol Chem 1986; 261: 15819-22. Cubellis MV, Andreasen PA, Ragno P, Mayer M, Dano K, Blasi F. Accessibility of receptor-bound urokinase to type-i plasminogen activator inhibitor. Proc Natl Acad Sci USA 1989; 86: 4828-32. Hajjar KA, Harpel PC, Jaffe EA, Nachman RL. Binding of plasminogen to cultured human endothelial cells. J Biol Chem 1986; 261: 11656-62. Miles LA, Plow EF. Receptor-mediated binding of the fibrinolytic components plasminogen and urokinase to peripheral blood cells. Thromb Haemost 1987; 58: 936-42. Burtin P, Fondaneche M-C. Receptor for plasmin on human carcinoma cells. J Natl Cancer Ins 1988; 89: 762-5. Plow EF, Freaney DE, Plescia J, Miles LA. The plasminogen system and cell surfaces: evidence of plasminogen and urokinase receptors on the same cell type. J Cell Biol 1986; 193: 2411-20. Beebe DP. Binding of tissue plasminogen activator to human umbilical vein endothelial cells. Thromb Res 1987; 46: 241-54. Pollanen J, Saksela O, Salonen E-M, et al. Distinct

CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

47.

48.

49. 50.

51. 52.

53. 54.

55.

56.

57.

58. 59.

60.

61. 62.

localization of urokinase-type plasminogen activator and its type-1 inhibitor under cultured human fibroblasts and sarcoma cells. J Cell Biol 1987; 104: 1085-96. Hebert CA, Baker JB. Linkage of extracellular plasminogen activator to the fibroblast cytoskeleton: colocalization of cell surface urokinase with vinculin. J Cell Biol 1988; 106: 1241-7. Thorsen S, Philips M. Plasminogen activator inhibitors. In: Castelino FJ, Gaffney PJ, Samama MM, Takada A, eds. Fundamental and clinical fibrinolysis. Pp. 83-98. Amsterdam: Elsevier, 1987. Sprengers ED, Kluft C. Plasminogen activator inhibitors. Blood 1987; 69: 381-7. Kruithoff EKO, Gudichet A, Bachmann F. Plasminogen activator inhibitor 1 and plasminogen activator inhibitor 2 in various disease states. Thromb Haemost 1988; 59: 7-12. Mignatti P, Robbins E, Rifkin DB. Tumor invasion through the human amniotic membrane: requirement for a proteinase cascade. Cell 1986; 47: 487-98. Reich R, Thompson EW, Iwamoto Y, et al. Effects of inhibitors of plasminogen activator, serine proteinases and collagenase IV on the invasion of basement membranes by metastatic cells. Cancer Res 1988; 48: 3307-12. Ossowski L. Plasminogen activator dependent pathways in the dissemination of human tumor cells in the chick embryo. Cell 1988; 52: 321-8. Mayer M, Lund LR, Riccio A, et al. Plasminogen activator inhibitor type-1 protein, mRNA and gene transcription are increased by phorbol esters in human rhabdomyosarcoma cells. J Biol Chem 1988; 263: 15688-93. Van Hinsberg VWM, Bertina RM, Van Wijngaarden A, Van Tilburg NH, Emeis JJ, Haverkate F. Activated protein C decreases plasminogen activatorinhibitor activity in endothelial ceU-conditioned medium. Blood 1985; 65: 444-51. Andreasen PA, Nielsen LS, Kristensen P, GrondahlHansen J, Skriver L, Dano K. Plasminogen activator inhibitor from human fibrosarcoma cells binds urokinase-type plasminogen activator, but not its proenzyme. J Biol Chem 1986; 261: 7644-51. Loskutoff DJ, Linders M, Keijer J, Veerman H, van Heerikhuizen H, Pannekoek H. Structure of the human plasminogen activator inhibitor 1 gene: nonrandom distribution of introns. Biochemistry 1987; 26: 3763-8. Travis J, Salvesen GS. H u m a n plasma proteinase inhibitors. Annu Rev Biochem 1983; 52: 655-709. Hekman CM, Loskutoff DJ. Endothelial cells produce a latent inhibitor of plasminogen activators that can be activated by denaturants. J Biol Chem 1985; 260: 11581-7. Hekman CM, Loskutoff DJ. Bovine plasminogen activator inhibitor 1: specificity determinations and comparison of the active, latent, and guanidineactivated forms. Biochemistry 1988; 27: 2911-8. Wohlwend A, Belin D, Vassali J-D. Plasminogen activator-specific inhibitors predueed by humanmonoeytes/ maerophages. J Exp Med 1987; 165: 320-39. Lund LR, Georg B. Nielsen LS, Mayer M, Dano K, Andreasen PA. Plasminogen activator inhibitor type 1: cell-specific and differentiation-induced expression and regulation in human cell lines, as deter-

207

MAYER

63.

64.

65.

66.

67.

68. 69.

70. 71.

72. 73.

74.

75.

76. 77. 78.

208

mined by enzyme-linked immunosorbent assay. Mol Cell Endocrin 1988; 60: 43-53. Kuiper J, Kamps AAM, Van Berkel TJC. Identification of the inhibitor of the plasminogen activator as the major protein secreted by endothelial rat liver cells. FEBS Lett 1989; 245: 229-34. Klinger KW, Winqvist R, Riccio A, et al. Plasminogen activator inhibitor type 1 gene is located at region q21.3-q22 of chromosome 7 and genetically linked with cystic fibrosis. Proc Natl Acad Sci USA 1987; 84: 8548-52. Medcalf RL, Van den Berg E, Schleuning W-D. Glucocorticoid-modulated gene expression of tissueand urinary-type plasminogen activator and plasminogen activator inhibitor 1 and 2. J Cell Biol 1988; 106: 971-8. Shleef RR, Bevilacqua MP, Sawdey M, Gimbrone, MA, Loskutoff DJ. Cytokine activation of vascular endothelium. Effect of tissue-type plasminogen activator and type 1 plasminogen activator inhibitor. J Biol Chem 1988; 263: 5797-803. Juhan-Vague I, Moerman B, De Cock F, Aillaud MF, Collen D. Plasma levels of a specific inhibitor of tissue-type plasminogen activator (and urokinase) in normal and pathological conditions. Thromb Res 1984; 33: 523-30. Sprengers ED. A sensitive assay, specific for endothelial cell type plasminogen activator inhibitor in blood plasma. Thromb Haemostas 1986; 55: 74-7. Vague P, Juhan-Vague I, Aillaud MF, et al. Correlation between blood fibrinolytic activity, plasminogen activator inhibitor level, plasma insulin level, and relative body weight in normal and obese subjects. Metabolism 1986; 35: 250-3. Wiman B, Csemicky G, Marsk L, Robbe H. The fast inhibitor of tissue plasminogen activator during pregnancy. Thromb Haemost 1984; 52: 124-6. Mimuro J, Loskutoff DJ. Purification of a protein from bovine plasma that binds to type 1 plasminogen activator inhibitor and prevents its interaction with extracellular matrix. Evidence that the protein is vitronectin. J Biol Chem 1989; 264: 936-9. Salonen E-M, Vaheri A, Pollanen J, et al. Interaction of plasminogen activator inhibitor (PAI-1) with vitronectin. J Biol Chem 1989; 264: 6339-43. Pollanen J, Hedman K, Nielsen LS, Dano K, Vaheri A. Ultrastructural localization of plasma membraneassociated urokinase-type plasminogen activator at focal contacts. J Cell Biol 1988; 106: 87-95. Antalis TM, Clark MA, Barnes T, et al. Cloning and expression of a cDNA coding for a human monocytederived plasminogen activator inhibitor. Proc Natl Acad Sci USA 1988; 85: 985-9. Ye RD, Ahren SM, Le Beau MM, Lebo RV, Sadler JE. Structure of the gene for human plasminogen activator inhibitor-2. The nearest mammalian homologue of chicken ovalbumin. J Biol Chem 1989; 264: 5495-502. Lecander I, Astedt B. Isolation of a new specific plasminogen activator inhibitor from pregnancy plasma. Br J Haematol 1986; 62: 221-8. Astedt B, Lecander I, Ny T. The placental type plasminogen activator inhibitor, PAI-2. Fibrinolysis 1987; 1: 203-8. Webb AC, Collins KL, Snyder SE, et al. Human monocyte Arg-Serpin cDNA. Sequence, chromosomal assignment, and homology to plasminogen activator inhibitor. J Exp Med 1987; 166: 77-94.

79. Chapman HA, Stone OL. A fibrinolytic inhibitor of human macrophages. Induction with endotoxin. A m Rev Respir Dis 1985; 132: 569-75. 80. Scott RW, Baker JB. Purification of human protease nexin. J Biol Chem 1983; 258: 10439-44. 81. Eaton DL, Scott RW, Baker JB. Purification of human fibroblast urokinase proenzyme and analysis of its regulation by proteases and protease nexin. J Biol Chem 1984; 259: 6241-7. 82. Hogstorp H, Saldeen T. Synthesis of a2-antiplasmin by rat liver cells. Thromb Res 1982; 28: 19-25. 83. Lottenberg R, Christensen U, Jackson CM, Coleman PL. In: Colowick S, Kaplan W, eds. Methods in enzymology. Vol. 80. Pp. 341-61. New York: Academic Press. 1981. 84. Mussoni L, Lawrence D, Loskutoff DJ. A direct, plasmin-independent assay for plasminogen activator. Thromb Res 1984; 34: 241-54. 85. Jespersen J, Astrup T. A study of the fibrin plate assay of fibrinolytic agents. Haemostasis 1983; 13: 301-15. 86. Lewis JG, Pizzo SV, Adams DO. A simple and sensitive assay employing stable reagents for quantification of plasminogen activator. A m J Clin Pathol 1981; 76: 403-9. 87. Mayer M, Finci Z, Chaouat M. Suppression of plasminogen activator activity by dexamethasone in cultured cardiac myocytes. J Mol Cell Cardiol 1986; 18: 1117-24. 88. Mayer M, Finci Z, Chaouat M, Sasson S. Plasminogen activator activity in differentiating rat myoblasts. Mol Cell Biochem 1986; 69: 147-54. 89. Mayer M, Yedgar S, Hurwitz A, Palti Z, Finci Z, Milwidsky A. Effect of viscous macromolecules on peritoneal plasminogen activator activity. A potential mechanism for their ability to reduce postoperative adhesion formation. A m J Obstet Gynecol 1988; 159: 957-63. 90. Wiman B, Mellbring G, Ranby M. Plasminogen activator release during venous stasis and exercise as determined by a new specific assay. Clin Chim Acta 1983; 127: 279-88. 91. Zaoui D, Le Fevre B, Magdlenat H, Bieth JG. A simple spectrophotometric assay of plasminogen activator: comparison with the fibrinolytic method. Clin Chim Acta 1984; 141: 101-9. 92. Chmielewska J, Wiman B. Determination of tissue plasminogen activator and its "fast" inhibitor in plasma. Clin Chem 1986; 32: 482-5. 93. Verheijen JH, Mullaart E, Chang GTG, Kluft C, Wijngaard G. A simple, sensitive spectrophotometric assay for extrinsic (tissue-type) plasminogen activator applicable to measurements in plasma. Thromb Haemost 1982; 48: 266-9. 94. Ranby M, Bergsdorf N, Nilsson T, Melbring G, Winblad B, Bucht G. Age-dependent tissue plasminogen activator concentrations in plasma studied by an improved enzyme linked immunosorbent assay. Clin Chem. 1986; 32: 2160-5. 95. Andreasen PA, Christensen TH, Huang J-Y, et al. Hormonal regulation of extracellular plasminogen activators and Mrs54000 plasminogen activator inhibitor in human neoplastic cell lines, studied with monoclonal antibodies. Mol Cell Endocrino11986; 45: 137-47. 96. Korninger C, Speiser W, Wojta J, Binder BR. Sandwich ELISA for tPA antigen employing a monoclonal antibody. Thromb Res 1986; 41: 527-35. CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

P L A S M I N O G E N ACTIVATION S Y S T E M

97. Chandler WL, Loo S-C, Nguyen SV, Schmer G, Stratton JR. Standardization of methods for measuring plasminogem activator inhibitor activity in human plasma. Clin Chem 1989; 35: 787-93. 98. Loskutoff DJ, Van Mourik JA, Erickson LA, Lawrence D. Detection of an unusually stable fibrinolytic inhibitor produced by bovine endothelial cells. Proc Natl Acad Sci USA 1983; 80: 2956-60. 99. Declerck PJ, Alessi MC, Verstreken M, Kruithof EKO, Juhan-Vague I, Collen D. Measurement of plasminogen activator inhibitor 1 in biological fluids with a murine monoclonal antibody-based enzymelinked immunosorbent assay. Blood 1988; 71: 220-5. 100. AIMS Trial Study Group. Effect ofintravenous APSAC on mortality after acute myocardial infarction: preliminary report of a placebo-controlled clinical trial. Lancet 1988; 1: 545-9. 101. Wilcox RG, v o n d e r Lippe G, Olsson CG, et al. Trial of tissue plasminogen activator for mortality reduction in acute myocardial infarction. Anglo-Scandinavian study of early thrombolysis (ASSET). Lancet 1988; 2: 525-30. 102. ISIS-2 Collaborative Group. Randomized trial of intravenous streptokinase, oral aspirin, both, or neither among 17,187 cases of suspected acute myocardial infarction: ISIS-2. Lancet 1988; 2: 349-60. 103. Marder VJ, Sherry S. Thrombolytic therapy: current status. N Eng J Med 1988; 318: 1512-20, 1585-94. 104. Loscalzo J, Braunwald E. Tissue plasminogen activator. N Eng J Med 1988; 319: 925-31. 105. Gurewich V. The sequential, complementary and synergistic activation of fibrin-bound plasminogen by tissue plasminogen activator and pro-urokinase. Fibrinolysis 1989; 3: 59-66. 106. Lijnen HR, Collen D. Molecular mechanisms of thrombolytic therapy. Haemostasis 1986; 16: suppl. 3, 3-15. 107. Mullertz S. Fibrinolysis. General aspects, characteristic features and perspectives. Fibrinolysis 1987; 1: 3-12. 108. Mullins DE, Rohrlich ST. The role of proteinases in cellular invasiveness. Biochim Biophys Acta 1983; 695: 177-214. 109. Liotta LA. Tumor invasion and metastases-role of the extracellular matrix. Cancer Res 1986; 46: 1-7. 110. Dano K, Andreasen PA, Grondahl-Hansen J, Kristensen P, Nielsen LS, Skriver L. Plasminogen activators, tissue degradation, and cancer. Adv Cancer Res 1985; 44: 140-266. 111. Hearing VJ, Law LW, Corti A, Apella E, Blasi F. Modulation of metastatic potential by cell surface urokinase of murine melanoma cells. Cancer Res 1988; 48: 1270-8. 112. Sullivan LM, Quigley JP. An anticatalytic monoclonal antibody to avian plasminogen activator: its effect on behavior of RSV-transformed chick fibroblasts. Cell 1986; 45: 905-15. 113. Tryggvason M, Hoyhtya M, Salo T. Proteolytic degradation of extracellular matrix in tumor invasion. Biochim Biophys Acta 1987; 907: 191-217. 114. Bar-Ner M, Mayer M, Schirrmacher V, Vlodavsky I. Involvement of both heparanase and plasminogen activator in lymphoma cell-mediated degradation of heparan sulfate in the subendothelial extracellular matrix. J Cell Physiol 1986; 128: 299-306. 115. Levin EG, Santell L. Association of a plasminogen activator inhibitor (PAL1) with the growth substratum and membrane of human endothelial cells. J CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

Cell Biol 1987; 105: 2543-9. 116. Coleman PL, Patel PD, Cwikel BJ, Rafferty UM, Snycer-Laszuk R, Gelehrter TD. Characterization of the dexamethasone-induced inhibitor of plasminogen activator in HTC hepatoma cells. J Biol Chem 1986; 261: 4352-7. 117. Sugawara Y, Takada Y, Yamamoto K, Takada A. Kinetic analysis of the enhancement of the activities of tPA induced by the presence of monoclonal antibody (C9-5). Thromb Res 1988; 50: 637-46. 118. Castellino FJ, Urano T, de-Serrano V, et al. Control of human plasminogen activation. Haemostasis 1988; 18 Suppl. 1: 15-23. 119. Higazi A, Mayer M. In vitro inhibition of urokinase by penicillins. Thromb Haemost 1989; 60: 305-7. 120. Scully MF, Ellis V, Watahiki Y, K a k k a r VV. Activation of pro-urokinase by plasmin: non-michaelian kinetics indicates a mechanism of negative cooperativity. Arch Biochem Biophys 1989; 268: 438--46. 121. Wiman B, Nilsson T, Cedergren B. Studies on a form of ~2-antiplasmin in plasma which does not interact with the lysine-binding sites in plasminogen. Thromb Res 1982; 28: 193-9. 122. McCabe RP, Evans CH. Plasminogen activator, fibronectin, lymphotoxin sensitivity, and natural skin reactivity relationships to guinea pig cell tumorigenicity. J Natl Cancer Inst 1982; 68: 329-35. 123. Ng R, Kellen JA, Wong ACH. Plasminogen activators as markers of tumor colonization potential. Invas Metast 1983; 3: 243-8. 124. Wilson L, Francis GE. Differentiation-linked secretion of urokinase and tissue plasminogen activator by normal human hemopoietic cells. J Exp Med 1987; 165: 1609-23. 125. Wilson EL, Jacobs P, Dowdle EB. The secretion of plasminogen activators by human myeloid leukemic cells in vitro. Blood 1983; 61: 568-74. 126. Francis CW, Marder VJ. Concepts of clot lysis. Annu Rev Med 1986; 37: 187-204. 127. Johan-Vague I, Valadier J, Alessi MC, et al. Deficient tPA release and elevated PA inhibitor levels in patients with spontaneous or recurrent deep venous thrombosis. Thromb Haemost 1987; 57: 67-72. 128. Stead NW, Bauer KA, Kinney TR, et al. Venous thrombosis in a family with defective release of vascular plasminogen activator and elevated plasma factor VIII/von Willebrand's factor. A m J Med 1983; 74: 33-9. 129. Aznar J, Estelles A, Vila V, Reganon E, Espana F, Villa P. Inherited fibrinolytic disorder due to an enhanced plasminogen activator level. Thromb Haemost 1984; 52: 196-200. 130. Kluft C, Verheijen JH, Jie AFH, et al. The postoperative fibrinolytic shut down: a rapidly reverting acute phase pattern for the fast-acting inhibitor of tissue-type plasminogen activator aider trauma. Scand J Clin Lab Invest 1985; 45: 605-10. 131. D'Angelo A, Kluft C, Verheijen JH, Rijken DC, Mozzi E, Manucci PM. Fibrinolytic shut-down a f a r surgery: impairment of the balance between tissuetype plasminogen activator and its specific inhibitor. Eur J Clin Invest 1985; 15: 308-12. 132. Paramo JA, Colucci M, Collen D. Plasminogen activator inhibitor in the blood of patients with coronary artery disease. Br J Med 1985; 291: 573-4. 133. Wiman B, Ljungberg B, Chimielewska J, Urden G, Blomback M, Johnsson H. The role of the fibrinolytic system in deep vein thrombosis. J Lab Clin Med

209

MAYER 1985; 105: 265-70. 134. Nilsson IM, Ljunguer H, Tengborn L. Two different mechanisms in patients with venous thrombosis and defective fibrinolysis: low concentration of plasminogen activator or increased concentration of plasminogen activator inhibitor. Br Med J 1985; 290: 14536. 135. Paramo JA, Alfaro MJ, Rocha E. Postoperative changes in the plasmatic level of tissue-type plasminogen activator and its fast acting inhibitor-relationship to deep vein thrombosis and influence of prophylaxis. Thromb Haemost 1985; 54: 713-6. 136. Paramo JA, De Boer A, Colucci M, Jonker JJC, Collen D. Plasminogen activator inhibitor (PA-inhibitor) activity in the blood of patients with deep vein thrombosis. Thromb Haemost 1985; 54: 725. 137. Hamsten A, Walldius G, Szamosi A, et al. Plasminogen activator inhibitor in plasma: risk factor for recurrent myocardial infarction. Lancet 1987; 1: 310. 138. Hamsten A, Wiman B, DeFaire U, Blomback M. Increased plasma levels of rapid inhibitor of tissue plasminogen activator in young survivors of myocardial infarction. N Eng J Med 1985; 313: 1557-63. 139. Kluft C, Preston FE, Malia RG, et al. Stanazololinduced changes in fibrinolysis and coagulation in healthy adults. Thromb Haemost 1984; 51: 157-64. 140. Rapaport E. Thrombolytic agents in acute myocardial infarction. N Eng J Med 1989; 320: 861-4. 141. Shafer KE, Santoro SA, Sobel BE, Jaffe AS. Monitoring activity of fibrinolytic agents. A therapeutic challenge. A m J Med 1984; 76: 879-86. 142. Peabody RA, Tsapogas MJ, Wu KT. Altered endogenous fibrinolysis and biochemical factors in atherosclerosis. Arch Surg 1974; 109: 309-13. 143. Stout RW. Diabetes and atherosclerosis. The role of insulin. Diabetologia 1979; 16: 141-50. 144. Almer L-O, Sundkvist G, Lilja B. Fibrinolytic activity, autonomic neuropathy, and circulation in diabetes mellitus. Diabetes 1983; 32: suppl. 2, 4-7. 145. Geiger M, Binder BR. Plasminogen activation in diabetes mellitus. Kinetic analysis of plasmin formation using components isolated from the plasma of diabetic patients. J Biol Chem 1984; 259: 2976-81. 146. Hall P, Tryon E, Nikolai F, Roberts RC. Functional activities and nonenzymatic glycosylation of plasma proteinase inhibitors in diabetes. Clin Chim Acta 1986; 160: 55-62. 147. Geiger M, Binder BR. Plasminogen activation in diabetes mellitus. Normalization of blood sugar levels improves impaired enzyme kinetics in vitro. Thromb Haemost 1985; 54: 413-4. 148. Medcalf RL, Richards RI, Crawford RJ, Hamilton JA. Suppression of urokinase-type plasminogen activator mRNA levels in human fibrosarcoma cells and synovial fibroblasts by anti-inflammatory glucocorticoids. EMBO J. 1986; 5: 2217-22. 149. Inman RD, Harpel PC. Alpha 2-plasmin inhibitorplasmin complexes in synovial fluid. J Rheumatol 1986; 13: 535-7. 150. Mochan E, Uhl J, Newton R. Interleukin 1 stimulation of synovial cell plasminogen activator production. J Rheumatol 1986; 13: 15-9. 151. Harpel PC, Gordon BR, Parker TS. Plasmin catalyzes binding of lipoprotein (a) to immobilized fibrinogen and fibrin. Proc Natl Acad Sci USA 1989; 86: 3847-51. 152. Ossowski L, Biegel D, Reich E. Mammary plasmino210

153. 154.

155.

156. 157.

158.

159. 160.

161.

162.

163.

164.

165.

166.

167.

168.

gen activator: correlation with involution, hormonal modulation and comparison between normal and neoplastic tissue. Cell 1979; 16: 929-40. Strickland S, Mahdavi V. The induction of differentiation in teratocarcinoma stem cells by retinoic acid. Cell 1978; 15: 393-403. Grondahl-Hansen J, Lund LR, Ralfkiaer E, Ottevanger V, Dano K. Urokinase- and tissue-type plasminogen activators in keratinocytes during wound re-epithelialization in vivo. Invest Dermatol 1988; 90: 790-5. Milwidsky A, Finci Z, Mayer M. Plasminogen activator activity and urokinase inhibitor activity in human amniotic fluid and fetal membranes. Clin Biochem 1988; 21: 301-6. Reich R, Miskin R, Tsafriri A. Follicular plasminogen activator: involvement in ovulation. Endocrinology 1985; 116: 516-21. Hsueh AJ, Liu Y-X, Cajander S, et al. Gonadotropinreleasing hormone induces ovulation in hypophysectomized rats: studies on ovarian tissue-type plasminogen activator activity, messenger ribonucleic acid content, and cellular localization. Endocrinology 1988; 122: 1486-95. Milwidsky A, Kaneti H, Finci Z, Laufer N, Tsafriri A, Mayer M. H u m a n follicular fluid protease and antiprotease activities: a suggested correlation with ability of oocytes to undergo in vitro fertilization. Fertil Steril 1989; 52: 274-80. Hettle JA, Waller EK, Fritz IB. Hormonal stimulation alters the type of plasminogem activator produced by sertoli cells. Biol Reprod 1986; 34: 895-904. Vihko KK, Toppari J, Pavrinen M. Stage-specific regulation of plasminogen activator secretion in the rat seminiferous epithelium. Endocrinology 1987; 120: 142-5. Gross JL, Moscatelli D, Rifkin DB. Increased capillary endothelial cell protease activity in response to angiogenic stimuli in vitro. Proc Natl Acad Sci USA 1983; 80: 2623-7. Vassali J-D, Dayer J-M, Wohlwend A, Belin D. Concomitant secretion of prourokinase and of plasminogen activator-specific inhibitor by cultured human monocytes-macrophages. J Exp Med 1984; 159: 1653-68. Rennie PS, Bouffard R, Bruchovsky N, Chen H. Increased activity of plasminogen activators during involution of the rat ventral prostate. Biochem J 1984; 221: 171-8. Degen JL, Estensen RD, Nagamine Y, Reich E. Induction and desensitization of plasminogen activator gene expression by tumor promoters. J Biol Chem 1985; 260: 12426-33. Mira-y-Lopez R, Reich E, Ossowski L. Modulation of plasminogen activator in rodent m a m m a r y tumors by hormones and other effectors. Cancer Res 1983; 43: 5467-77. Mira-y-Lopez R, Reich E, Stolfi R, Martin DS, Ossowski L. Coordinate inhibition of plasminogen activator and tumor growth by hydrocortisone in mouse m a m m a r y carcinoma. Cancer Res 1985; 45: 2270-6. Busso N, Belin D, Failly-Crepin C, Vassali J-D. Glucocorticoid modulation of plasminogen activators and of one of their inhibitors in the human mammary carcinoma cell line MDA-MB-231. Cancer Res 1987; 47: 364-70. Peltz SW, Katzenellenbogen BS, Kneifel MA, Mangel WF. Plasminogen activators in tissues of the CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

PLASMINOGEN ACTIVATION SYSTEM

169. 170.

171.

172.

173.

174.

175. 176.

177.

immature and estrogen-stimulated rat uterus and in uterine luminal fluid: characterization and properties. Endocrinology 1983; 112: 890-7. Van Hinsberg VWM. Regulation of the sysnthesis and secretion of plasminogen activators by endothelial cells. Haemostasis 1988; 18: 307-27. Heaton JH, Nebes VL, O'Dell LG, Morris SM, Gelehrter TD. Glucocorticoid and cyclic nucleotide regulation ofplasminogen activator and plasminogen activatorinhibitor gene expression in primary cultures of rat hepatocytes. Mol Endocrinol 1989; 3: 185-92. Ashino-Fuse H, Opdenakker G, Fuse A, Billiau A. Mechanism of the stimulatory effect of phorbol 12myristate 13-acetate on cellular production of plasminogen activator. Proc Soc Exp Biol Med 1984; 176: 109-18. Coleman PL, Patel PD, Cwikel BJ, Rafferty UM, Snycer-Laszuk R, Gelehrter TD. Characterization of the dexamethasone-induced inhibitor of plasminogen activator in HTC hepatoma cells. J Biol Chem 1986; 261: 4352-7. Andreasen PA, Pyke C, Riccio A, et al. Plasminogen activator inhibitor type 1 biosynthesis and mRNA level are increased by dexamethasone in human fibrosarcoma cells. Mol Cell Biol 1987; 7: 3021-5. Georg B, Helseth E, Lund LR, et al. Tumor necrosis factor-~ regulates mRNA for urokinase-type plasminogen activator and type-1 plasminogen activator inhibitor in human neoplastic cell lines. Mol Cell Endocrinol 1989; 61: 87-96. Gelehrter TD, Snycer-Laszuk R. Thrombin induction of plasminogen activator-inhibitor in cultured human endothelial cells. J Clin Invest 1986; 77: 165-9. Lund LR, Riccio A, Andreasen PA, et al. Transforming growth factor-~ is a strong and fast acting positive regulator of the level of type-1 plasminogen activator inhibitor mRNA in WI-38 human lung fibroblasts. EMBO J 1987; 6: 1281-6. Crutchley DJ, Conanan LB. Endotoxin induction of

CLINICAL BIOCHEMISTRY, VOLUME 23, JUNE 1990

178.

179.

180.

181.

182.

183.

184.

185.

an inhibitor of plasminogen activator in bovine pulmonary artery endothelial cells. J Biol Chem 1986; 261: 154-9. Colucci M, Paramo JA, Collen D. Generation in plasma of a fast inhibitor of plasminogen activator in response to endotoxin stimulation. J Clin Invest 1985; 75: 818-24. Gramse M, Breviario F, Pintucci G, et al. Enhancement by interleukin-1 (IL-1) of plasminogen activator inhibitor (PA-1) activity in cultured human endothelial cells. Biochem Biophys Res Commun 1986; 139: 720-7. Nachman RL, Hajjar KA, Silverstein RL, Dinarello CA. Interleukin 1 induces endothelial cell synthesis of plasminogen activator inhibitor. J Exp Med 1986; 163: 1595-1600. Bevilacqua MP, Schleef RR, Gimbrone MA, Loskutoff DJ. Regulation of the fibrinolytic system of cultured human vascular endothelium by interleukin 1. J Clin Invest 1986; 78: 587-91. Westerhausen DR, Hopkins WE, Sobel BE, Billadello JJ. Transcriptional regulation of type 1 plasminogen activator inhibitor in HEP G2 cells by epidermal growth factor. J Cell Biochem 1989; suppl. 13E: 204 (Abstr.). Kruithoff EKO, Vassali J-D, Schleuning W-D, Mattaliano RJ, Bachmann F. Purification and characterization of a plasminogen activator inhibitor from the histiocytic lymphoma cell line U-937. J Biol Chem 1986; 261: 11207-13. Schleuning W-D, Medcalf RL, Hession C, Rothenbuhler R, Shaw A, Kruithof EKO. Plasminogen activator inhibitor 2: regulation ofgene transcription during phorbol ester-mediated differentiation of U-937 human histiocytic lymphoma cells. Mol Cell Biol 1987; 7: 4564-7. Wun T-C, Capuano A. Initiation and regulation of fibrinolysis in human plasma at the plasminogen activator level. Blood 1987; 69: 1354-62.

211

Biochemical and biological aspects of the plasminogen activation system.

Plasminogen activators (PAs) are specific proteolytic enzymes which convert the inactive proenzyme plasminogen to plasmin. The plasmin formed is a pot...
2MB Sizes 0 Downloads 0 Views