THE KALLIKREIN-KININ system was first recognized as a plasma and tissue proteolytic system responsible for the liberation of the vasoactive, proinflammatory mediator, bradykinin (BK).1 BK, a nonapeptide released from kininogens by kallikreins, could reproduce many of the characteristics of an inflammatory state, such as changes in local blood pressure, edema, and pain, resulting in vasodilation and increased microvessel permeability. In 1975, three individuals were described with deficiency of high molecular weight kininogen (HK), a precursor of BK, all of whom had a prolonged activated partial thromboplastin time (APTT), a surface-activated coagulation protein screening test.2-4 Despite the fact that none of these individuals had a hemorrhagic state, studies on the plasma kallikrein-kinin system focused on defining the procoagulant property of HK. In fact, it was already known that deficiency of the two zymogens, factor XII and prekallikrein, required for the enzymatic cleavage of HK, also did not lead to bleeding. These plasma proteins together were grouped as the contact system because they required contact with artificial, negatively charged surfaces for zymogen activation. Over the last 20 years, these proteins have been shown to have little influence on hemostasis. However, examination of their molecular, biochemical, biologic, and physiologic properties has shown that these proteins interact with a number of physiologic and pathophysiologic systems. Cloning and delineation of their structure-function relationships have shown new activities of these proteins such as protease inhibition, antithrombin function, and antiadhesive properties. Their specific interactions with biologic membranes of endothelial cells, platelets, neutrophils, and monocytes indicate that assembly and activation of this system takes place in a physiologic milieu, independent of negatively charged surfaces. In fact, it is correct to say that the so-called elusive physiologic, negatively charged surface for contact system activation is actually the assembly of these proteins on cell membranes. In vivo, a negatively charged surface is not needed for activation. One may argue that the term contact activation is a misnomer to describe this system. The proteins of the plasma contact system have anticoagulant, profibrinolytic, antiadhesive, and proinflammatory functions. This review presents a revitalized view of the contact system as a physiologic mediator of vascular biology and inflammatory reactions. We will first examine the current structure-function knowledge of each of the proteins of the system: HK, prekallikrein, and factor XII. We will next describe how this system assembles on cell membranes. The participation of these proteins in various biologic activities (eg, blood pressure regulation, inhibition of thrombin activation of cells, cellular fibrinolysis, and antiadhesion) then will be characterized in terms of their assembly and activation on cell membranes. Furthermore, we will describe both clinical examples and experimental models in which this system is activated. It is the goal of this review to clarify the contributions of this system to physiologic and pathophysiologic reactions of vascular biology. Last, this presentation will point to possible new therapeutic strategies to treat various diseases arising out of the knowledge of this system in physiologic and pathophysiologic states.

High Molecular Weight Kininogen (Williams, Fitzgerald Factor)

Gene expression and regulation. The two forms of plasma kininogens, HK and low molecular weight kininogen (LK), are the products of a single gene.5,6 This gene maps to 3q26-qter, the location of the homologous α2HS-glycoprotein and histidine-rich glycoprotein.7-9 The single kininogen gene of 11 exons consisting of 27 kb produces a unique mRNA for HK and LK by alternative splicing (Fig 1).6 HK and LK share the coding region of the first nine exons, a part of exon 10 containing the BK sequence, and the first 12 amino acids after the carboxy-terminal sequence of BK. Exon 11 codes for a unique 4-kD light chain of LK. The complete exon 10 contains the full coding sequence for the unique 56-kD light chain of HK. A novel mechanism for alternative RNA processing has been characterized in the rat kininogen gene.10 Splicing efficiency is controlled by the interaction of U1 small nuclear ribonucleoproteins and the U1 small nuclear RNA (snRNA)-complementary repetitive sequences of the kininogen pre-mRNA. The mRNA for LK and HK are 1.7 and 3.5 kb, respectively.

Fig. 1.

The domain structure of the kininogens. The kininogens are produced by one gene with 11 exons (E1-E11). E1-E3 codes for domain 1 (D1) on both HK (high molecular weight kininogen) and LK (low molecular weight kininogen). Parts of domain 1 inhibit atrial naturetic factor. E4-E6 codes for domain 2 (D2), which has papain and unique calpain inhibitory sequences. E7-E9 codes for domain 3 (D3), which has papain inhibitory sequences. Domain 4 (D4) is coded by part of E10; it is the bradykinin sequence on kininogens and the first 12 amino acids of the light chains of HK and LK. The remainder of E10 codes for HK's light chain, which consists of domain 5 (D5H ) and domain 6 (D6H ). D5H is an artificial surface binding region; D6H has the prekallikrein and factor XI binding regions. Domains 3, 4, and 5 on HK also participate in cell binding. E11 codes for the remainder of the unique light chain of LK (D5L ).

Fig. 1.

The domain structure of the kininogens. The kininogens are produced by one gene with 11 exons (E1-E11). E1-E3 codes for domain 1 (D1) on both HK (high molecular weight kininogen) and LK (low molecular weight kininogen). Parts of domain 1 inhibit atrial naturetic factor. E4-E6 codes for domain 2 (D2), which has papain and unique calpain inhibitory sequences. E7-E9 codes for domain 3 (D3), which has papain inhibitory sequences. Domain 4 (D4) is coded by part of E10; it is the bradykinin sequence on kininogens and the first 12 amino acids of the light chains of HK and LK. The remainder of E10 codes for HK's light chain, which consists of domain 5 (D5H ) and domain 6 (D6H ). D5H is an artificial surface binding region; D6H has the prekallikrein and factor XI binding regions. Domains 3, 4, and 5 on HK also participate in cell binding. E11 codes for the remainder of the unique light chain of LK (D5L ).

Close modal

The molecular basis for one example of homozygous total kininogen deficiency, Williams trait, has been determined.11 A C to T transition at nucleotide 587 occurred, changing a CGA (Arg) codon to TGA (Stop) mutation in exon 5 and resulting in prevention of synthesis of both HK and LK.11 The phenotype of this defect is similar to that seen in Brown-Norway, Katholiek strain rats that have absent plasma kininogens, but the defect in the rats is due to a single point mutation, Ala163 to Thr, which results in defective secretion from the liver.12 Little is known about what regulates gene expression of kininogens. In the rat, ovariectomy results in a reduction of kininogen transcripts in the liver, whereas estrogens increase kininogen mRNA levels.13 This result is consistent with the clinical observation that HK concentrations increase in pregnancy.14 In contrast, progesterone treatment reduced kininogen gene expression, resulting in a slight reduction of plasma kininogen levels.15 Murine fibroblasts synthesize and secrete kininogens in response to cyclic-AMP, forskolin, prostaglandin E2 , and tumor necrosis factor α.15 Similarly, tumor necrosis factor α has been recognized to increase kininogen expression in HEP G2 cells.16 Little else is known to influence kininogen levels, only because this aspect of kininogens has not been studied extensively.

Protein chemistry and structure of the kininogens. The two mRNAs of the kininogens code for two separate proteins. LK is a 66-kD β-globulin with a plasma concentration of 160 μg/mL (2.4 μmol/L) and an isoelectric point of 4.7.17,18 HK is a 120-kD α-globulin with a plasma concentration of 80 μg/mL (0.67 μmol/L) and an isoelectric point of 4.3.18,19 Human liver is a source for cDNA for both kininogens,5,6 but human umbilical vein endothelial cells have been shown to contain HK mRNA and to synthesize the protein.20 Kininogen antigen also has been found in platelets, granulocytes, renal tubular cells, and skin.19-24 LK, until its cloning, was also known as an α1 -cysteine protease inhibitor.25 Both HK and LK are composed of globular units. LK gel filters at 66 kD and behaves as a true globular protein; HK, although 120 kD, gel filters at 220 kD, indicating a high axial ratio. Physical evidence for HK being a complex of globular units was obtained by electronmicroscopy studies.26 On electron microscopy, HK appeared to be a linear array of three linked centralized globular regions, with the two ends thinly connected.26 Cleavage of HK by plasma kallikrein leads to a striking change in conformation in HK. The central globular region is separated after bradykinin liberation and rearranged with the cysteine protease inhibitory region opposite the prekallikrein binding region.26 The regions of kininogens are divided into domains (Fig 1). Separating these domains are serine protease sensitive regions.27-29 As will be discussed below, contiguity of certain domains are important for some biologic functions of kininogens such as calpain inhibition and HK and LK binding to endothelial cells.30-32 Alternatively, proteolytic cleavage of HK unmasks a new function, ie, its cell antiadhesive activity.33 Of course, the major activity of kininogens, which is to deliver bradykinin, is programmed disruption of the protein, because bradykinin is not active as a biologic peptide unless liberated from its precursor.

Domain structure of kininogens. The kininogens are proteins composed of multiple domains, each with associated activities (Fig 1). Binding of kininogen to its cell receptors facilitates bradykinin liberation in a circumscribed environment in which the peptide can bind to bradykinin receptors and influence the local cellular milieu. Thus, one can view each function of the domains of the kininogens as participating in the whole protein's kinin delivery activity. The kininogens, in general, can be divided into three portions: the heavy chain that is common to both HK and LK, the bradykinin moiety, and the light chains that, as already stated, are unique to HK and LK, respectively (Fig 1). Domains 1 through 3 comprise kininogens' heavy chain. Domain 4 is the bradykinin region. Domain 5 for LK (D5L ) is its unique 4-kD light chain. Domains 5 and 6 of HK (D5H or D6H ) are unique to this protein and comprise its light chain.

Little is known about the function of domain 1 except to note that it has a low-affinity binding calcium binding site whose role is unknown.34 Although calcium ions are important for phorbol 12-myristate 13-acetate upregulation of LK and heavy chain binding to endothelial cells,35 there is no good evidence that calcium ions participate in HK binding to cells,31,36 contrary to other laboratories' work.37,38 Recent evidence also indicates that a peptide from domain 1 inhibits atrial naturetic peptide.39 Domains 2 and 3 contain the highly conserved amino acid sequence, QVVAG, found in cysteine protease inhibitors (Fig 2).27 Both LK and HK are potent, tight-binding, reversible cysteine protease inhibitors with Kis of 2 and 0.5 nmol/L, respectively, of platelet calpain.18,40 Kininogens' calpain inhibitory region is exclusively found on domain 228,30,40,41, whereas papain and cathepsin L are effectively inhibited by regions on both domains 2 and 3.27,30,42,43 Computer three-dimensional models of domain 2 were constructed using x-ray crystallographic coordinates of cystatin, which is 50% identical to domains 2 and 3.30 Peptides from domain 2 of HK were selected and air-oxidized to form disulfide-bonded loops. A peptide containing Q170VVAG174 blocked HK inhibition of calpain and thus functioned as a binding site (Fig 2). Another peptide (C211-C229) C-terminal to this peptide was a direct inhibitor of calpain (IC50 = 35 μmol/L). The two regions probably form a continuous binding site on the three-dimensional structure of kininogens (Fig 2). A third peptide (V128-L138), N-terminal to the QVVAG region, inhibited papain, but not calpain, indicating that the inhibitory sites on domain 2 for these two cysteine proteases are not identical (Fig 2). In contrast, the optimal inhibition of cathepsin B and H requires three loops of domain 3 (Fig 2).43 Although an inhibitor of cysteine proteases, kininogens are also substrates of this class of enzyme when there is molar excess of enzyme to inhibitor.18,44,45 Because kininogens are extracellular or within granules in platelets and granulocytes, it has been unclear how they interact with cellular cysteine proteases that, for the most part, are internal membrane or cytosolic in location.19,21,46 However, when platelets are activated, calpain translocates to the external membrane in which it could be inhibited by plasma or externalized platelet α-granule HK.45-47 

Fig. 2.

The structure of high molecular weight kininogen. An amino acid sequence diagram of high molecular weight kininogen. A circle with thin vertical lines represents a papain inhibitory domain. A circle with thin horizontal lines represents a calpain inhibitory domain. A circle with a solid background represents a cell surface binding domain. A circle with thick vertical lines represents overlapping papain inhibitory activity and cell surface binding activity. A circle with thick horizontal lines represents bradykinin. A circle with a blank backround represents the factor XI binding domain. A circle with a shaded background represents overlapping prekallikrein and factor XI binding domain.

Fig. 2.

The structure of high molecular weight kininogen. An amino acid sequence diagram of high molecular weight kininogen. A circle with thin vertical lines represents a papain inhibitory domain. A circle with thin horizontal lines represents a calpain inhibitory domain. A circle with a solid background represents a cell surface binding domain. A circle with thick vertical lines represents overlapping papain inhibitory activity and cell surface binding activity. A circle with thick horizontal lines represents bradykinin. A circle with a blank backround represents the factor XI binding domain. A circle with a shaded background represents overlapping prekallikrein and factor XI binding domain.

Close modal

Domain 3 of kininogens has other functions. The finding that LK and its isolated heavy chain bind to platelets and endothelial cells indicates that there is a cell binding region on kininogens' heavy chain.35,48,49 This point was confirmed by direct studies using isolated and recombinant domain 3 that contained the heavy chain cell binding region on platelets50 and neutrophils.51 Using a computerized model of domain 3 also based on the structure of crystallized cystatin,52 the sequential amino acid structure of domain 3 was drawn to show three surface-exposed regions: a disulfide loop connecting it to domain 2 and two hairpin loops (Fig 2). The cysteine protease inhibitory region of domain 3 consists of portions of these three surface-exposed loops. Using synthetic peptides of these surface-exposed regions, K244ICVGCPRDIP254 (KIC11), N276ATFYFKIDNVKKARVQVVAGKKYFI301 (NAT26), and L331DCNAEVYVVPWEKKIYPTVNC-QPLGM357 (LDC27), studies were performed to determine if they inhibit HK binding to endothelial cells. KIC11, NAT26, and LDC27 inhibited biotin-HK binding to endothelial cells with IC50 of 1,000, 258, and 60 μmol/L, respectively. The minimal sequence in LDC27 to inhibit binding was 13 amino acids, C333NAEVYVVPWEKK345 (IC50 = 113 μmol/L).53 Because papain blocked HK binding to endothelial cells, the cysteine protease inhibitory site overlaps with the cell binding site on domain 3.53 Thus, the last 27 amino acids of domain 3, which are contiguous to domain 4, the bradykinin region, are an endothelial cell binding site. Thrombospondin (TSP), a platelet α-granule protein secreted upon platelet stimulation, also binds to HK both to a site on the heavy chain requiring calcium ions and to the light chain independent of calcium ions.54 TSP's interaction with kininogens' heavy chain may be on domain 3 overlapping the KIC11 sequence.54 

The last function ascribed to domain 3 was kininogens' α-thrombin inhibitory activity.48,50,55 Isolated domain 3, prepared by tryptic digestion of LK in solution, inhibited α-thrombin–induced platelet activation.50 The thrombin inhibitory region was not the same as the platelet binding region because one monoclonal antibody (MoAb), which did not block cell binding, neutralized HK's ability to inhibit α-thrombin's activation of platelets.50 Furthermore, the α-thrombin inhibitory region on kininogens was not one of the three cell binding regions, KIC11, NAT26, or LDC27.53,56,57 Two other distinct sequences, one from domain 3 and another contiguous with domain 3 on domain 4, respectively, are capable of inhibiting thrombin-induced platelet activation by different mechanisms. Kunapuli et al57 expressed domain 3 in Escherichia coli, G235-M357. The recombinant polypeptide inhibited thrombin-induced aggregation of platelets with an IC50 of 4 μmol/L. It should be noted that this sequence, unlike the tryptic digest of LK, does not include any part of domain 4. The protein coded by exon 7, G235-Q292, showed an IC50 of 13.4 μmol/L, and a recombinant peptide of 23 amino acids, K270-Q292, showed an IC50 of 30 μmol/L. Finally, a synthetic heptapeptide located on domain 3, L271-A277 (LNAENNA), was the minimal sequence to inhibit α-thrombin–induced platelet aggregation (IC50 = 65 μmol/L). As will be described below, this sequence competes for thrombin binding to platelets by mimicking a GPIb sequence on platelets for binding thrombin.

Alternatively, Hasan et al56 indicate that the kininogens' thrombin inhibitory activity previously ascribed to domain 3,50 prepared by proteolytic cleavage, is really domain 4, or the kinin moiety remaining attached to the C-terminus of domain 3. When pure or plasma HK is cleaved by plasma kallikrein on an artificial surface, bradykinin is liberated from its parent protein in three ways.58,59 The first cleavage yields a nicked kininogen composed of two disulfide-linked 64- and 56-kD chains. The second cleavage yields bradykinin (0.9 kD) and an intermediate kinin-free protein of approximately similar molecular weight to nicked HK. The third cleavage results in a stable, kinin-free protein composed of two disulfide-linked 64- and 46-kD chains. However, when kininogens are cleaved in solution without a surface, this sequence does not necessarily occur and bradykinin can remain attached to kininogens' heavy or light chain.60 Because isolated domain 3 was prepared by proteolytic cleavage in solution, we examined both trypsin-cleaved LK and domain 3 prepared by tryptic digestion and found that the bradykinin moiety remained attached to LK's heavy chain and isolated domain 3.32 Investigations were next performed to determine if bradykinin, analogs of bradykinin, and its breakdown products block α-thrombin–induced platelets activation. In experiments to be described below, all of these domain 4 fragments were shown to be inhibitors of α-thrombin–induced platelet aggregation by preventing α-thrombin from cleaving its cloned receptor (PAR1).56 

Certainly, domain 4, the bradykinin region, has many functions assigned to this nanopeptide in addition to its newest function, α-thrombin inhibition.56 In the liberation of bradykinin, HK is a better substrate of plasma kallikrein and LK is a better substrate of tissue kallikrein. However, both kininogens are substrates to both forms of kallikrein. Factor XIIa cleaves HK similarly to plasma kallikrein.61 Factor XIa initially cleaves HK into 76- and 46-kD bands. Upon prolonged exposure to factor XIa, the 46-kD light chain of HK is proteolyzed into smaller, inactive fragments.62 Elastase treatment of LK renders the protein a better substrate of plasma kallikrein to liberate bradykinin and Met-Lys-bradykinin,63 although it destroys HK's procoagulant activity. Cathepsin D inactivates kininogens' cysteine protease inhibitory activity.64 One last function of domain 4 is to serve as a cell binding region.65 The carboxy terminal portion of bradykinin and the amino terminal portion of kininogen's common light chain participate as a low-affinity (kd = 1 mmol/L) binding site to endothelial cells. The importance of the domain 4 cell binding region is not its isolated affinity to the cell surface, but its ability to hold kininogens in the proper conformation for optimal cell binding.26 For example, intact HK binds to endothelial cells maintained at 37°C with a kd of 7 nmol/L and 1 × 107 molecules/cell versus kinin-free kininogen, which binds to endothelial cells maintained at 4°C with a kd of 30 nmol/L and 1 to 2.6 × 106 molecules/cell.31 These different data for intact or kinin-free HK's interaction with biologic surfaces are not surprising considering the major change in the shape of HK that occurs when it is cleaved on an artificial surface.26 

LK's light chain is 4 kD and consists of one domain (D5L ). Its function is not known. HK's light chain is 56 kD and consists of two domains, domains 5 (D5H ) and 6. D5H serves as an additional cell binding site on platelets, granulocytes, and endothelial cells.35,49,51,66 Two areas of D5H were found to participate in cell binding.67 One is on the amino terminal end of the domain and consisted of sequences G402KEQGHTRRHDWGHEKQRK420 (GKE19) and H421NLGHGHKHERDQGHGHQRGH441 (HNL21) (Fig 2). These peptides inhibit biotin-HK binding with IC50 of 792 and 215 μmol/L, respectively.67 The other region is on the carboxy terminal region of D5H and is subsumed in the region of two overlapping peptides H479KHGHGHGKHKNKGKKNGKH498 (HKH20) and H471VLDHGHKHKHGHGHGKHKNKGKK494 (HVL24) that inhibit HK binding with IC50 of 0.23 and 0.8 μmol/L, respectively.67 Preliminary evidence suggests that the region responsible for binding to neutrophils on D5 is localized to H420-H458, similar to HNL21.68 Independent of its cell binding region, D5H has been recognized as HK's artificial surface binding region.28,69,70 D5H's histidine- and glycine-rich regions have the ability to bind to anionic surfaces, zinc, and heparin.69-72 Using an MoAb that blocks HK clotting and binding of cleaved HK to anionic surfaces,28 a 7.3-kD peptide was isolated on an immunoaffinity column that was identified by N-terminal analysis as H441-K497.72 This 57 amino acid peptide inhibited coagulant activity and had the ability to bind to anionic surfaces with an IC50 of 30 μmol/L. D5H contains two histidine- and glycine-rich regions, one on its carboxy terminal side, which was also rich in lysine (H457-K502), similar to HKH20, and the other on its amino terminal side (K420-H458), similar to HNL21. Using a deletion mutagenesis strategy on D5H , the anionic surface binding region was found to be associated with both histidine-glycine–rich regions of D5H .70 Either region was able to support coagulant activity provided it was associated with D6.70 This question was examined further using synthetic peptides.67 Peptides HKH20 and HVL24, which are found to be its high-affinity cell binding regions on the carboxy terminal side of D5H , are also found to inhibit the procoagulant activity of HK.67 No other peptides from D5H , including HNL21, have this property. Furthermore, a polyclonal antibody reared to HKH20 is able to prolong the procoagulant activity of HK in plasma.67 These data indicate that the endothelial cell and neutrophil cell binding regions and the artificial surface binding region on HK are contained within the same highly conserved region of D5H . Furthermore, the endothelial cell and artificial surface binding regions are overlapping.67 Peptide HKH20 and its parent HK have the additional ability to interact with M protein on Streptococcus pyogenes.73 It is of interest that the highest affinity cell binding site for D5H turns out to be the artificial surface binding site. Efforts by many investigators over the last two decades to characterize HK binding to artificial surfaces indicated the location of HK's cell binding site. Last, when HK is bradykinin free, the residual kinin-free kininogen has the ability to prevent the adhesive interaction of vitronectin with tumor cells, endothelial cells, platelets, and monocytes.33 This property is much weaker in intact, nonproteolyzed HK. This result was anticipated by the finding that, after the liberation of bradykinin from HK, the resulting kinin-free kininogen binds much more tightly to anionic surfaces than does the uncleaved HK.74 

HK's domain 6 has a prekallikrein (S565-K595) and factor XI binding site (P556-M613) (Fig 2).75-77 The affinity of prekallikrein for its binding site on the light chain of HK is about 17 nmol/L.78,79 The prekallikrein and factor XI binding site consists of a 31-residue sequence that contains predominantly β-turn elements.80 Although the 30 amino acid region (S565-K595) was shown to be sufficient for binding, more recent studies show that an N-terminally and C-terminally truncated 27-mer (W569-K595) has the essential structural elements for prekallikrein binding.81,82 HK's procoagulant activity is dependent on two activities: (1) the ability to bind to anionic surfaces via D5H and (2) the ability to bind prekallikrein and factor XI to domain 6.28 Inhibition of either interaction with MoAbs directed to these regions will inhibit HK's procoagulant activity.28,83,84 HK's domain 6 serves as the acceptor protein for factor XI and prekallikrein binding to platelets, neutrophils, and endothelial cells.37,85,86 As will be seen below, prekallikrein binding to bound HK initiates a sequence of events that leads to prekallikrein activation on biologic surfaces independent of factor XII activation.

Prekallikrein (Fletcher Factor)

Prekallikrein (PK) is produced by a single gene that maps to chromosome 4.87 PK's gene structure is similar to that of factor XI.88 Its mRNA codes for a 371 amino acid heavy chain and a 248 amino acid light chain that are held together by a disulfide bond (Fig 3).88 The amino acid sequence of PK has 58% homology to factor XI.88 The protein has four tandem repeats in the amino-terminal portion of the molecule due to the linking of the first and sixth, second and fifth, and third and fourth half cysteines residues present in each repeat (Fig 3). This arrangement results in four groups of 90 or 91 amino acids that are arranged in so-called apple domains.89,90 These same structures have been described in factor XI, suggesting a common ancestor genic duplication event for plasma prekallikrein and factor XI.87,91 

Fig. 3.

The structure of prekallikrein. The letters A1 through A4 represent the apple domains of prekallikrein's heavy chain. The notation Factor XIIa and arrow at arginine371 represent the factor XIIa activation site on prekallikrein. Histidine415 , aspartic acid464 , and serine559 represent kallikrein's catalytic active site. A circle with a shaded background represents the regions involved in binding to high molecular weight kininogen. Adapted from Chung et al.89 

Fig. 3.

The structure of prekallikrein. The letters A1 through A4 represent the apple domains of prekallikrein's heavy chain. The notation Factor XIIa and arrow at arginine371 represent the factor XIIa activation site on prekallikrein. Histidine415 , aspartic acid464 , and serine559 represent kallikrein's catalytic active site. A circle with a shaded background represents the regions involved in binding to high molecular weight kininogen. Adapted from Chung et al.89 

Close modal

In plasma, PK appears as a doublet of 85 and 88 kD, whether or not the protein has undergone reduction.92,93 In plasma, PK is a fast γ-globulin (isolelectric point = 8.5 to 9.0) with a circulating concentration in blood estimated at 35 to 50 μg/mL (0.41 to 0.56 μmol/L).94,95 Human liver has been shown to be a source for PK cDNA.89 In liver disease, plasma PK is decreased.94 Women on oral contraceptives have increased PK levels, but women in their second and third trimester of pregnancy do not.14,94 When PK is activated to kallikrein (α-kallikrein) by either factor XIIa or factor XIIf, the protein on reduced sodium dodecyl sulfate (SDS) gel electrophoresis has two subunits: a heavy chain of approximately 52 kD and two light chains variants of approximately 36 and 33 kD.92,93 The active site of kallikrein is contained within its light chain because this region incorporates tritiated diisopropyl fluorophosphate in a covalent linkage with serine559 .96 Histidine415 and asparatic acid464 comprise the other two amino acids involved in catalytic activity (Fig 3). Prolonged incubation of kallikrein with itself results in autodigestion of its heavy chain into 33- and 20-kD bands as seen on reduced SDS gel electrophoresis to yield a form termed β-kallikrein.97 These cleavages occur through the tandem repeats in the heavy chain and result in a protein that cleaves HK more slowly and fails to activate neutrophils or induce their secretion of elastase.97,98 Nonreduced SDS gel electrophoresis of artificial, negatively charged surface activation of plasma results in the appearance of kallikrein in complex with α2macroglobulin (α2M) and C1 inhibitor as well as the appearance of a 50-kD prekallikrein/kallikrein fragment containing a portion of the native protein's heavy chain.14 At least 75% of PK circulates bound, noncovalently, to HK.99 The binding regions on PK for HK are on apple domains 1 (F56-G86) and 4 (K266-G295) (Fig 3).100-104 The prekallikrein binding regions for factor XII are localized in apple domains 3 and 4, but the specific sequence has not been delinated.100 

The in vitro conversion of human plasma PK to kallikrein, its active form, is catalyzed by activated factor XII, on a surface augmented by HK, or by Hageman factor fragment (βFXIIa) in the fluid phase.96 In the absence of factor XII, prekallikrein will not become activated on an artificial surface. It is because of this finding that this system is called the contact system. A single bond (Arg371 -Ile372 ) is split, generating a heavy chain of 371 amino acids still linked to a light chain by a single disulfide bridge without a change in molecular weight. On the endothelial cell surface, this cleavage occurs in the absence of factor XII when PK is bound to HK.86 The light chain of kallikrein reacts with protease inhibitors, principally α2M and C1 inhibitor (C1-INH). C1-INH forms a 1:1 stoichiometric complex with kallikrein,105-109 resulting in loss of proteolytic and amidolytic activity. HK protects kallikrein from inhibition by C1-INH and α2M in a purified system,109,110 suggesting a mechanism of substrate (HK) protection of the enzyme (kallikrein) from active site-directed protease inhibitors. α2M inhibits the kinin-forming activity but only partially inhibits the amidolytic activity of kallikrein108 by forming a covalent complex. Although C1 inhibitor and α2M account for an equal amount of kallikrein inhibition in plasma, C1 inhibitor in plasma acts more rapidly than α2M.111 Antithrombin III also inhibits kallikrein, but it does so slowly, even in the presence of heparin.112 In the presence of HK, heparin, which binds to HK,71,72 significantly accelerates the inhibition of kallikrein by antithrombin. Protein C inhibitor has also been recognized to be a potent inhibitor of kallikrein.113,114 The major protein substrates of plasma kallikrein are factor XII, HK, and prourokinase.115,116 

Factor XII (Hageman Factor)

Factor XII is produced by a single gene that maps to chromosome 5.117,118 The gene for factor XII is 12 kb and is composed of 13 introns and 14 exons.119 By both the complementary DNA (cDNA) and DNA sequence, factor XII has multiple domains with extensive sequence homology with regions of tissue-type plasminogen activator (tPA; the epidermal growth factor [EGF ]-like region and the kringle region) and fibronectin (Fig 4).119-121 The factor XII intron/exon gene is similar in organization to the serine protease family of tPA and urokinase-type plasminogen activator (uPA) genes, but is different from most other coagulation protein genes.119 Its 2.4-kb mRNA codes for a 596 amino acid, single chain β-globulin with a molecular mass of 80 to 90 kD and an isoelectric point of 6.1 to 6.5 (Fig 4).122 Its concentration in plasma is estimated to be 30 μg/mL (0.375 μmol/L; range, 15 to 47 μg/mL).123,124 Human liver has been shown to be a source for factor XII DNA120 and cultured rat hepatocytes synthesize factor XII.125 In humans, estrogens administered to postmenopausal women and pregnant women elevate plasma levels of factor XII and its expression is enhanced in isolated livers of estrogen- and prolactin-treated rats.126-128 Rat liver DNA has been shown to have a functional estrogen regulatory element contained in its 5′ untranslated region that is modulated by 17β-estradiol.129 Factor XII, which contains an EGF domain, enhances HepG2 cell proliferation and thymidine and leucine incorporation, suggesting that it is a mitogen for these cells.130 In fact, factor XII, through its EGF domain, functions as a mitogen and stimulates a signal transduction pathway by a mitogen-induced protein kinase.131 This activity is independent of activated factor XII's proteolytic activity.

Fig. 4.

The structure of factor XII. Proteolysis at arginines 334, 343, and 353 (see arrows) results in activated factor XII (β-factor XIIa). The catalytic triad of factor XIIa consists of histidine393 , aspartic acid442 , and serine544 . A circle with a shaded background represents the artificial surface binding domains on factor XII's heavy chain. A circle with horizontal lines represent two of factor XII's zinc binding domains. Adapted from Cool and MacGillivray.119 

Fig. 4.

The structure of factor XII. Proteolysis at arginines 334, 343, and 353 (see arrows) results in activated factor XII (β-factor XIIa). The catalytic triad of factor XIIa consists of histidine393 , aspartic acid442 , and serine544 . A circle with a shaded background represents the artificial surface binding domains on factor XII's heavy chain. A circle with horizontal lines represent two of factor XII's zinc binding domains. Adapted from Cool and MacGillivray.119 

Close modal

Factor XII can be divided into two regions, a heavy chain and a light chain. The heavy chain contains two artificial surface binding regions, one at the distal amino terminal end (I1-C28) and another on its fibronectin type I region (T134-R153) (Fig 4).132,133 Recent studies using recombinant deletion mutants of factor XII confirmed these findings and also indicated that a third region on factor XII's heavy chain, on the second EGF-like or kringle domain (P313-R334, L344-R353), also participated in artificial surface binding (Fig 4).134 Upon contact with negatively charged surfaces, FXII is autoactivated (solid-phase activation).135 Both the binding to the surface and the cleavage during autoactivation result in distinct, defined conformational changes.136 Plasma proteinases, including plasma kallikrein and plasmin, activate factor XII (FXII) to FXIIa (αFXIIa), cleaving the bond connecting Arg353 -Val354 and generating a two-chain molecule composed of a heavy chain (353 residues) and a light chain (243 residues), held together by a disulfide bond.120 The light chain of FXIIa is a typical serine proteinase containing the canonical Asp442 , His393 , and Ser554 and is the site for inhibition by its major plasma inhibitor, C1-inhibitor.137 Hageman factor fragments or FXII fragments (FXIIf, βFXIIa) (Mr = 30 kD) are produced by further proteolytic cleavage, resulting in a chain of 243 residues expressing catalytic activity attached to a fragment of the former heavy chain by a single disulfide bond. Defects in the light chain of factor XII result in disorders of the enzymatic activity of the protein. Coagulation factor XII Washington DC has a Cys571 -to-Ser substitution that results in complete loss of procoagulant activity.138 Coagulation factor XII Bern is a protein that, when kallikrein-cleaved, is unable to activate factor XI or prekallikrein.139 Contact activation arises from the activation of factor XII. Factor XII can be activated by contact with negatively charged surfaces or by the addition of a protease that produced enzymatic cleavage. These two mechanisms have been referred to as solid- and fluid-phase activation, respectively.140 

The activation of factor XII that arises from binding with negatively charged surfaces140-143 is termed autoactivation.144-149 Some evidence suggests that Zn2+ binding to factor XII induces a conformation change that makes the protein more susceptible for development of enzymic activity when associated with negatively charged surfaces.150-152 There are four zinc binding sites, two of which have been identified (H40-H44 and H78-H82).153 Although there are large number of candidate physiologic negatively charged surfaces that in vitro can be associated with factor XII autoactivation, the concept of autoactivation itself has never been a sufficiently convincing mechanism to explain activation of factor XII and associated contact system activation in vivo. Alternative mechanisms have been searched for factor XII activation in vivo. A rabbit endothelial cell activator of factor XII has been described, but there is no corresponding example in humans.154 Our own studies indicate that incubation of zymogen factor XII with human umbilical vein endothelial cells does not result in human factor XII activation (unpublished data). However, assembly of PK bound to HK on human umbilical vein endothelial cells results in PK activation independent of factor XII by a cell-associated thiolprotease.86 Furthermore, factor XII activation by this pathway can occur. In the absence of prekallikrein, factor XII does not activate on endothelial cells in a purified system or in plasma (unpublished data).

Enzymatic activation of factor XII gives rise to successively smaller proteins, each with the same active site serine site. Activation of zymogen factor XII by plasma kallikrein, trypsin, or plasmin results in an enzyme with a decreasing size, a decrease in its surface-binding properties, and a decrease in its coagulant activity. There are two major forms of activated Hageman factor: factor XIIa (αXIIa), an 80-kD protein consisting of two disulfide-linked polypeptide chains, and factor XIIf (Hageman factor fragments, HFf, βXIIa), a 28- to 30-kD fragment derived from factor XIIa.155-159 β-Factor XIIa results from cleavage at arginines 334, 343, and 353.120 The 80-kD form of activated factor XII has the ability to bind to negatively charged surfaces133,134 and activate factor XI. The 28- to 30-kD enzymatic form of factor XII has no surface-binding properties but retains its ability to activate prekallikrein and C1.140,160,161 

The major plasma protease inhibitor of activated factor XIIa and XIIf is C1 inhibitor, accounting for greater than 90% of the inhibition of these proteases in plasma.162-165 C1 inhibitor will bind both proteins and irreversibly inactivate them. When associated with a kaolin surface, factor XIIa is protected from C1 inhibitor inactivation.166 Antithrombin III has some inhibitory activity on factor XIIa.167,168 Plasminogen activator inhibitor-1 (PAI-1) also inhibits factor XIIa.169 Endothelial cells may also produce a protein that impairs factor XII activation, but not its coagulant or amidolytic activity once formed.170 

A major impediment to appreciate the contact system is the pervasive notion that the system has no biologic relevance because it is entirely activated on artificial surfaces. Although most studies to date only describe activation of this system on artificial surfaces and much work has been performed to describe physiologic, negatively charged surfaces (eg, acidic phospholipids, cholesterol sulfate, sulfatides, gout crystals, etc), none has been convincing as a single, unifying in vivo activator of this system. The physiologic, negatively charged surface for contact system activation is actually the assembly of these proteins on biologic surfaces, ie, cell membranes. In the protected milieu of cell membranes, we have now shown that the assembly of contact proteins on endothelial cell membranes leads to a multiprotein complex that results in prekallikrein activation independent of activated factor XII.86 This mechanism will be discussed in a later section. Detailed investigations of the proteins of the contact system interacting with cells have led to this current hypothesis as to how this system is physiologically active. Although there are some individual cell differences, we will first discuss the common features of contact protein expression and interaction with cells in the intravascular compartment.

The pivotal protein for contact system assembly on cell membranes is HK. In addition to being contained within platelets, granulocytes, and endothelial cells, unoccupied binding sites for HK exists on each of these cells.19-21,36-38, 45,171,172 Why each of these cells contain kininogens and also have unoccupied binding sites for them is not known. In platelets, less than 8% of total platelet HK is HK tightly bound to the platelet membrane.19,45 Upon platelet activation, 40% of total platelet HK is secreted and another 40% of the total becomes expressed upon the activated platelet membrane.45 The total platelet contribution to plasma HK is only 0.23%.19,173 The local concentration of HK on or about the activated platelet membrane may exceed 10 times the plasma concentration of this protein because platelets excrete their granule contents by exocytosis.19,45 

The majority of granulocyte-associated HK appears to be exogenous HK tightly bound and nonexchangeable with the granulocyte surface.174 Granulocytes have the ability to assemble all of the proteins of the contact system.174 Elastase liberated from granulocytes proteolyzes cell-bound HK.175 Initial investigations suggested that human umbilical vein endothelial cells were able to internalize HK.20,172 However, more recent detailed investigations indicate that there is no mechanism for HK internalization by endothelial cells.31 The difference in the amount of HK associated with the endothelial cell membrane when cells are maintained at 4°C versus 37°C is that, at the higher temperature, there is increased expression of kininogen binding sites.20,31,35,172,176 

There are characteristic features of kininogens binding to all cells. First, kininogen binding to cells has an absolute requirement for Zn2+.20,21,36,38,171,172 The requirement for Zn2+ is probably not limited to mediate HK binding to the cells by its zinc binding region of domain 5.69,72 LK binding to platelets and endothelial cells also has an absolute requirement for Zn2+. These data indicate that Zn2+ is necessary for the expression of the kininogen binding site, putative receptor.35,48 Although some investigators have suggested that calcium is a cofactor for binding to endothelial cells and platelets, our investigations show that it does not influence HK binding to unstimulated platelets, endothelial cells, or granulocytes.21,31,36 However, calcium was a requirement for maximal upregulation of LK or isolated heavy chain binding to endothelial cells after stimulation with phorbol esters.35 When HK or LK binds to platelets, granulocytes, or endothelial cells, the affinity of binding are the similar (Table 1). Because the affinity of HK binding to cells in the intravascular compartment is between 7 and 52 nmol/L and the plasma concentration of HK is 670 nmol/L, we can postulate that all kininogen binding sites in the intravascular compartment are saturated in vivo. The number of binding sites for the kininogens on cells in the intravascular compartment varies with the cell type. Platelets have approximately 1,000 binding sites/cell; granulocytes have approximately 50,000 sites/cell; and endothelial cells have approximately 1,000,000 sites/cell when chilled to 4°C and approximately 10,000,000 sites/cell when maintained at 37°C (Table 1).20,21,31,35,36,50 

Table 1.

Kininogen Expression on Cells in the Intravascular Compartment

Cell Typekd (nmol/L)*No. of Sites
Platelets 
125I-HK 15 ± 4 911 ± 239 
125I-LK 27 ± 2 647 ± 147 
125I-D3 39 ± 8 1,227 ± 404 
Granulocytes 
125I-HK 10 ± 1.3 4.8 × 104 
Endothelial cells 
125I-HK at 4°C 52 ± 13 9.3 × 105 
125I-LK at 4°C 43 ± 8 9.7 × 105 
Biotin-HK at 4°C 46 ± 8 2.6 × 106 
Biotin-HK at 37°C 7 ± 3 1.0 × 107 
Cell Typekd (nmol/L)*No. of Sites
Platelets 
125I-HK 15 ± 4 911 ± 239 
125I-LK 27 ± 2 647 ± 147 
125I-D3 39 ± 8 1,227 ± 404 
Granulocytes 
125I-HK 10 ± 1.3 4.8 × 104 
Endothelial cells 
125I-HK at 4°C 52 ± 13 9.3 × 105 
125I-LK at 4°C 43 ± 8 9.7 × 105 
Biotin-HK at 4°C 46 ± 8 2.6 × 106 
Biotin-HK at 37°C 7 ± 3 1.0 × 107 
*

Values presented were determined by direct binding studies.

Values presented represent the mean ± SD.

The expression of kininogens on cell membranes is a complex process. As indicated above in previous sections, there appear to be multiple regions on kininogens that allow them to interact with its various cellular receptors. The first information that such was the case was the finding that HK binds to platelets, endothelial cells, and granulocytes by regions on their heavy and light chains.35,49,51,66 HK actually has three domains that fit into the putative kininogen receptor(s) on endothelial cells.65 The interaction sites between HK and its putative receptor may be multiple locations: 3 in domain 3, 1 in domain 4, and 2 in domain 5.53,65,67 Clearly, the sequence of peptide LDC27 from domain 3 and HKH20 from domain 5 are the highest affinity binding regions on HK for endothelial cells.53,67 It is important to appreciate that the binding of even a low-affinity sequence from domain 4, for example, will block whole HK from binding to endothelial cells.65 This information suggests that HK and, presumably, LK have a very tight fit into its binding site(s), putative receptor(s). In fact, because the Ki and Kd calculated from binding studies for HK, LK, and all of their subunits are the same, the two chains of kininogens do not bind to cells in an optimal manner.66,177 This kind of noncooperative interaction is characterized by a loss of entropy on binding and suggests that whole HK bends to fit into its binding site, putative receptor.177 In support of this notion, when bradykinin is liberated from HK, kinin-free HK binds to endothelial cells with lower affinity and number of binding sites.31,65 Likewise, when LK is cleaved between domains 1 and 2 such that there is a change in the conformation of the LK, there is decreased LK binding to endothelial cells compared with intact LK.32 These changes in the biology of HK expression on cell membranes when bradykinin is removed from the protein are, in retrospect, predictable from the major conformational changes that take place between HK and kinin-free kininogen as shown in functional characteristics74 in electronmicroscopy26 and documented by circular dichroism.178 

The kininogen binding site, putative receptor on endothelial cells appears to be a structure that can be regulated. First, treatment of endothelial cells with metabolic inhibitors to anaerobic and aerobic metabolism and the hexose monophosphate shunt abolish the ability of HK to bind to the cells.31 Cycloheximide has no effect on HK binding to endothelial cells. Second, temperature or the bradykinin sequence in kininogens contributes to the level of kininogen binding to endothelial cells.31,65 Third, bradykinin treatment of endothelial cells results in increased HK and LK binding and this pathway is mediated by protein kinase C and the endothelial cell B1 bradykinin receptor.35 Fourth, heavy chain and LK have a Ca2+ requirement for phorbol 12-myristate 13-acetate 4-0-methyl ether upregulation of their endothelial cell binding site, whereas HK does not.35 Fifth, angiotensin-converting enzyme inhibitors potentiate the effect of bradykinin on upregulating the HK binding site on endothelial cells.35 Last, when HK binds to endothelial cells, it initiates a series of events that allow for an endothelial cell- or matrix-associated enzyme to activate prekallikrein bound to HK.86 Thus, bradykinin upregulates kininogen binding on endothelial cells and kininogens can influence bradykinin formation.35,86 These data indicate that this system is tightly controlled in an autocrine-like manner.

The combined data described above indicate that there should be a physiochemical receptor(s) for kininogens on blood and endothelial cells. Recent evidence proposes a number of candidate proteins to be the kininogen receptor(s). Antibody inhibition studies suggest that Mac-1 (CD11b/18) may be an HK binding site on granulocytes.51 Fibrinogen has been shown to be a noncompetitive inhibitor of HK binding to granulocytes and ADP-stimulated platelets.175 HK could bind directly to CD11b/18 on granulocytes or could interact with a receptor complexed to that integrin (see below). Herwald et al180 have isolated on a HK affinity column from EA.hy926 cells, a human umbilical vein endothelial cell line,179 a 33-kD protein that was identified as gC1qR. gC1qR is a known C1 receptor protein181 that only binds HK and peptides from domain 5, but not LK or binding peptides from domain 3. Furthermore, its ability to bind HK does not require Zn2+, although other workers claim that Zn2+ is required for ligand blots.182 Moreover, only a small portion of total endothelial cell gC1qR is found on the external membrane of endothelial cells.183 These data indicate that gC1qR cannot explain all of the characteristics of the kininogen receptor. Factor XII blocks HK binding to qC1qR.180 These data support the previous finding that factor XII partially blocks HK binding to endothelial cells.184 The kininogen binding protein just described may form part of a multiprotein receptor complex to explain the features of HK and LK binding to cells. Recently, preliminary evidence has been presented that HKa also binds to the urokinase receptor on endothelial cells.185 An antibody to domain 2/3 of the urokinase receptor completely inhibits HKa binding to endothelial cells, as does vitronectin, a ligand for this receptor domain. Soluble urokinase receptor markedly inhibits the binding of HKa and forms a zinc-dependent complex with it in a cell-free system. The finding that integrins are tightly associated with the urokinase receptor186 and can enhance the binding of ligands to domain 2/3 of the urokinase receptor could be relevant to the interaction of kininogens with neutrophils, which display both integrins and the urokinase receptor. Recent evidence indicates that HKa binds directly to cells transfected with Mac-1 and to purified Mac-1.186a The interaction of the urokinase receptor with CD11b/18 could be a potential pathway by which kininogen binding could signal within cells. However, because platelets do not express the urokinase receptor, this candidate binding site also cannot be the major kininogen receptor on all cells. Recent evidence indicates that cytokeratin 1 is an additional kininogen (HK and LK) binding site on endothelial cells, platelets, and granulocytes.187 Kininogen binding to cytokeratin 1 requires Zn2+ and all cell binding domains of kininogens interact with it. gC1qR and suPAR block HK binding to cytokeratin, suggesting that these proteins participate in a multiprotein assembly on endothelial cells. These data along with the recent finding that cytokeratin 8 is a cellular plasminogen receptor suggest that cytokeratins may represent a new class of presentation receptors on cells.188,189 Full characterization of the multiprotein kininogen receptor complex is the next challenge in this field.

On endothelial cells and platelets, kininogen binding modulates activation of the contact system. Platelet and endothelial cell bound HK is protected from activation by exogenous plasma kallikrein.66,190 Moreover, HK serves as the binding site or receptor for factor XI and prekallikrein on platelets and endothelial cells.37,85,86,191 No evidence exists to date to indicate that platelet-associated factor XI is activated to factor XIa in any favorable fashion.192 However, prekallikrein bound to HK on platelets or endothelial cells can result in its activation to kallikrein by a factor XIIa-dependent85,193 or independent86 mechanism. The factor XII-independent prekallikrein activation mechanism is due to a membrane- or matrix-associated thiolprotease whose activity is regulated by HK binding.86 Both situations result in the generation of bradykinin.86 Thus, cell membrane assembly of contact proteins through binding can result in a complex that can be activated through physiologic mechanisms to result in bradykinin liberation and the kinin-dependent activities.

In addition to the general characteristics of contact proteins interacting with cells of the intravascular compartment as described above, there are some unique protein-cell interactions as well. Kallikrein, but not PK, is chemotactic for neutrophils.194 Exposure of neutrophils to concentrations of kallikrein capable of eliciting chemotaxis increased aerobic glycolysis and activity of the hexose-monophosphate shunt.194 In the presence of calcium, neutrophils aggregate in response to kallikrein.195 This interaction is associated with stimulation of the respiratory burst in neutrophils, as indicated by an increase in oxygen uptake.195 Kallikrein also induces neutrophils to release human neutrophil elastase from their azurophilic granules196 and primes neutrophils for superoxide production.197 

In plasma, human neutrophils release elastase during blood coagulation,198 but neutrophils resuspended in either PK- or FXII-deficient plasma release less than one-third of the amount of elastase released in normal human plasma.196 A skin window technique that assesses the in vivo chemotaxis of leukocytes in response to tissue or microvascular injury shows a significant impairment in chemotaxis in FXII and PK-deficient patients.199 This result suggests that both kallikrein and FXIIa are important in the release of elastase from neutrophils in plasma. In addition, kallikrein induces an in vitro release of elastase from neutrophils in a concentration-dependent fashion that requires the presence of both the active site of kallikrein (on its light chain) and an intact heavy chain.200 The requirement for an uncleaved heavy chain can be explained by the requirement for both apple 1 and apple 4 sequences for binding of kallikrein to HK on neutrophils.104,105 Kallikrein formation occurring in human sepsis and experimental arthritis and enterocolitis (see below) would also recruit neutrophils to participate in the body defenses. FXIIa has also been shown to cause neutrophil aggregation201 and degranulation (release of elastase). FXIIf will not stimulate neutrophils, and, thus, a domain on the heavy chain is required. However, the catalytic activity of FXIIa is required because the active site inhibitors, D-Pro-Phe-Arg-CH2Cl and corn trypsin inhibitor, both abolish the reaction.

Factor XIIa can decrease the number of FcγR1 (Ig) receptors on monocytes without affecting its affinity. This interaction requires the heavy chain, but, in contrast to the effect of FXIIa on neutrophils, does not require the catalytic apparatus of the light chain.202 The site on FXII responsible for the downregulation of FcγR1 may be within the N-terminal 18 amino acids,203 and this decrease could impair the clearance of immune complexes. Toossi et al204 have found that factor XII induced monocyte synthesis and secretion of interleukin-1 (IL-1) and IL-6. These investigators found that lipopolysaccharide-stimulated secretion of these interleukins is also potentiated by factor XII.

The simple fact that a deficiency of HK, prekallikrein, and factor XII prolongs artificial surface-activated clotting without being associated with bleeding has obfusicated understanding the role of this system in vivo. The absence of hemostatic states associated with these proteins does not lessen their importance. The dicotomy between abnormal surface-activated screening laboratory tests for bleeding states and in vivo hemostasis should give us caution in interpreting laboratory tests as predictors of bleeding. Independent of its lack of effect on hemostasis, contact system activation modulates vascular biology. The multidomain kininogens have a number of biologic activities either within the intact protein or becoming manifest when the intact protein is proteolyzed by kallikreins or activated factor XII. This system is a potent local regulator of blood pressure through bradykinin delivery. It also has both selective antithrombin and profibrinolytic activity. Lastly, the cleaving of HK unmasks antiadhesive properties of the protein as well.

Bradykinin Delivery

The first and most enduring function of the plasma kininogens is the delivery of bradykinin, a potent biologically active peptide.1 In many ways, kininogens and bradykinin, an activation peptide from domain 4, contribute to vessel patency, increased blood flow, and anti-thrombotic/profibrinolytic activities (Table 2). Bradykinin itself is a potent stimulator of endothelial cell prostacyclin synthesis; an inhibitor of platelet function,205,206 superoxide formation,207 and tissue plasminogen activator release; and a stimulator of plasminogen activation,208,209 nitric oxide formation,210 and endothelial cell-dependent smooth muscle hyperpolarization factor formation.211 Furthermore, bradykinin, through its ability to stimulate NO and cGMP formation in endothelial cells, provides a major stimulus to prevent subendothelial smooth muscle proliferation.212,213 In the presence of an intact endothelium, kinins appear to prevent vascular smooth muscle growth and proliferation.214,215 Alternatively, when vessels are injured, bradykinin stimulates protein kinase C and subsequently MAP kinases that can result in vascular smooth muscle growth and proliferation.215-217 Thus, in an intact vessel, the sum of bradykinin activities is to keep blood flowing and vessels patent; in the absence of endothelium, bradykinin stimulates repair of vessels that could lead to smooth muscle proliferation and intimal hypertrophy.

Table 2.

Kininogens' Antithrombin, Antiadhesive, and Profibrinolytic Activities

DomainActivity
Bradykinin Stimulates prostacyclin formation 
Bradykinin Stimulates NO formation 
Bradykinin Stimulates superoxide formation 
Bradykinin Selectively stimulates tissue plasminogen activator secretion 
RPPGF Prevents α-thrombin from cleaving its receptor (PAR1) 
Domain 1 Inhibits atrial naturetic factor 
Domain 2 Prevents calpain-related platelet aggregation 
Domain 3 Prevents α-thrombin binding to platelets and endothelial cells 
Domain 5 Prevents cells from sticking to artificial surfaces 
Domain 5 Displaces fibrinogen from surfaces and cells 
Domain 6 Prekallikrein and factor XI receptor on endothelial cells and neutrophils 
DomainActivity
Bradykinin Stimulates prostacyclin formation 
Bradykinin Stimulates NO formation 
Bradykinin Stimulates superoxide formation 
Bradykinin Selectively stimulates tissue plasminogen activator secretion 
RPPGF Prevents α-thrombin from cleaving its receptor (PAR1) 
Domain 1 Inhibits atrial naturetic factor 
Domain 2 Prevents calpain-related platelet aggregation 
Domain 3 Prevents α-thrombin binding to platelets and endothelial cells 
Domain 5 Prevents cells from sticking to artificial surfaces 
Domain 5 Displaces fibrinogen from surfaces and cells 
Domain 6 Prekallikrein and factor XI receptor on endothelial cells and neutrophils 

Bradykinin effects its changes in the intravascular compartment by binding to at least two receptors, the B1 and B2 receptors.218,219 Both of these receptors are G-coupled; thus, binding of bradykinin stimulates cellular signal transduction. Increased bradykinin results in increased cellular stimulation. Blocking of the B2 receptor with an antagonist Hoe 140 (D-Arg,[Hyp3,Thi5,D-Tic7,Oic8]-bradykinin) in developing rats results in higher blood pressures, heart rates, and body weights than controls.220 The in vivo modulation of bradykinin levels by angiotensin converting enzyme (ACE) inhibitors is believed to be the basis for the cardioprotective attributes of these agents.221,222 ACE inhibitors induced NO and prostacyclin formation in cultured bovine endothelial cells and protect isolated perfused hearts from ischemia.223 The effects of ACE inhibitors to elevate NO and protect isolated ischemic hearts was abolished by the B2 receptor antagonist, Hoe 140.224 ACE inhibitor treatment in spontaneously hypertensive rats prevented the development of hypertension and left ventricular hypertrophy and Hoe 140 blocks these effects.225,226 These data in animals have been extended to humans, in whom ACE inhibitors have also been shown to be protective against myocardial infarction by increasing myocardial blood flow and decreasing ischemic changes. Although these cardioprotective effects of ACE inhibitors may be sufficiently explained alone by bradykinin's effect on vasculature, recent information that this peptide and its breakdown products are also selective inhibitors of α-thrombin could contribute as well (see below).56 

Blood Pressure Regulation

Although much is known about the physiologic effects of bradykinin and how its stimulates its response in cells, little is known on what regulates its liberation from kininogens by kallikreins. What regulates prekallikrein and tissue kallikrein activation on the vascular endothelium is not known. Regulation of these zymogen's activation is important because they directly modulate bradykinin liberation, which, in turn, has a direct effect on blood pressure in vivo. Dextran sulfate activation of rat plasma in vivo induces arterial hypotension, which can be blocked by a B2 receptor antagonist.227 Transgenic mice overexpressing tissue kallikrein are hypotensive.228 Intramuscular delivery of rat kallikrein-binding protein reverses hypertension in transgenic mice overexpressing human tissue kallikrein.229 Rat kallikrein binding protein or kallistatin is the cognate SERPIN of tissue kallikrein.230-232 Gene delivery of tissue kallikrein reduced mean blood pressure of spontaneous hypertensive rats and this inhibition was blocked by kallistatin.233,234 These molecular genetic studies directly indicate that presumed tissue kallikrein induced bradykinin liberation directly modifies local and, if sufficiently diffuse, systemic blood pressure regulation.

Thrombin Inhibition

In addition to the salutory effects of kinins to maintain vessel patency, bradykinin's precursor proteins, the kininogens, have been shown to selectively inhibit α-thrombin–induced platelet activation. There are at least three mechanisms by which kininogens influence α-thrombin–induced platelet and endothelial cell activation48,50,235 (Table 2). The first mechanism is an indirect one probably mediated by kininogen's ability to inhibit platelet calpain. When α-thrombin activates platelets, cytosolic or internal membrane-associated platelet calpain translocates to the activated platelet surface.46,47 Externalized platelet calpain is able to proteolyze platelet surface membrane glycoproteins such as glycoprotein Ib.236 Platelet calpain also proteolyzes a putative platelet ADP receptor that exposes the platelet fibrinogen receptor and thus allows for platelet aggregation.235,237 Thus, inhibition of externalized platelet calpain by leupeptin or HK, ie, inhibitors of calpains, results in inhibition of α-thrombin–mediated platelet aggregation by preventing fibrinogen binding.55,235 These data have been used to develop a group of compounds modeled after kininogens' domain 2 that prevent α- and γ-thrombin–induced platelet aggregation without interfering with other platelet agonists and thrombin-induced intracellular platelet activation.238 Selective thrombin-induced platelet aggregation inhibitors can be developed by making peptides modeled after kininogens' domain 2.

Additional studies suggested that there are more mechanisms by which kininogens inhibit α-thrombin–induced platelet activation. HK and LK were found to inhibit α-thrombin–induced platelet aggregation and secretion.48,56 Because α-thrombin–induced platelet secretion is independent of and occurs before platelet aggregation, kininogens must interfere with α-thrombin–induced platelet activation by other mechanisms than just inhibition of calpain-related platelet aggregation.239 HK, LK, and D3 were found to noncompetitively inhibit α-thrombin, but not Phe-Pro-Arg-chloromethylketone–treated thrombin, from binding to the platelet high-affinity site and endothelial cells.31,48,50,56,240 This finding was one explanation of how all platelet activation by α-thrombin could be blocked by large molecular mass proteins such as HK and LK. Additional studies have recognized other mechanisms for inhibition.

Kunapuli et al57 found that recombinant domain 3 (containing no residues of domain 4) inhibited thrombin-induced aggregation of platelets with only twofold less affinity than purified HK, indicating that at least one site for inhibition does reside in domain 3 (Fig 5). The minimal α-thrombin inhibitory sequence was Leu271 -Ala277 .57 Leu271 -Ala277 did not inhibit platelet aggregation by ADP or collagen. It did not inhibit thrombin's amidolytic or clotting activity. Leu271 -Ala277 also failed to inhibit platelet shape change and it did not inhibit SFLLRN from aggregating platelets. Bradford et al241 have obtained evidence that Leu271 -Ala277 and K270-Q292 inhibited thrombin-induced platelet activation at low thrombin concentrations by inhibiting the binding of thrombin to GPIb-IX-V complex. Furthermore, antibodies to and ligands of GPIbα inhibited HK binding to platelets and HK inhibited binding of antibodies to GPIbα to platelets. Moreover, domain 3 peptides directly inhibited high-affinity 125I–α-thrombin binding to platelets. Finally, HK inhibited binding of thrombin to fibroblasts transfected with GPIb-IX-V. These findings suggested that domain 3 peptides may block α-thrombin binding to its high-affinity site on GPIbα. The sequence NAEN appears in HK, domain 3 peptides, and the ligand binding domain of GPIbα. It is possible that HK's domain 3 may mimic this high-affinity binding site for thrombin. These findings do not necessarily imply that binding of thrombin to GPIbα itself results in platelet activation. Rather, GPIbα may serve to present thrombin to the G-protein–linked cloned thrombin receptor, thus lowering the concentration of thrombin necessary to cleave the latter receptor. Kininogens by blocking this interaction would then modulate thrombin-induced platelet activation.

Fig. 5.

Kininogens' thrombin inhibitory domains. A circle with a solid background represents thrombin inhibitory activity. A circle with a shaded background represents a cell membrane binding region. A circle with diagonal lines represents overlapping papain inhibitory activity and cell membrane binding activity.

Fig. 5.

Kininogens' thrombin inhibitory domains. A circle with a solid background represents thrombin inhibitory activity. A circle with a shaded background represents a cell membrane binding region. A circle with diagonal lines represents overlapping papain inhibitory activity and cell membrane binding activity.

Close modal

A third mechanism of α-thrombin inhibition has been described by Hasan et al.56 Kininogens and peptides derived from it actually inhibit α-thrombin–induced platelet activation by blocking the enzyme's ability to cleave the cloned thrombin receptor (PAR1).56 In work already described above, purified domain 3 prepared from proteolytic cleavage actually had domain 4 attached to it.32,56 Peptides from domain 4, BK, and related sequences were found to inhibit α-thrombin–induced platelet activation (Fig 5). Although large molecular mass HK, LK, and D3 inhibited α-thrombin binding to platelets, isolated domain 4, ie, bradykinin (RPPGFSPFR) and MKRPPGFSPFRSSRIG, did not inhibit binding.48,50,56 These data indicated that another mechanism is operative for these peptides to block α-thrombin–induced platelet activation. Like the parent proteins HK and LK, domain 4 peptides did not inhibit α-thrombin's ability to cleave a tripeptide substrate or clot fibrinogen, suggesting that these peptides did not interact with α-thrombin's active site or anion binding exosite.31,48,50,56 Moreover, like HK and LK, these peptides were not substrates of α-thrombin and they did not form complexes with α-thrombin.31,48,56 Domain 4 peptides did not block ADP-, collagen-, or U46619-induced platelet aggregation in vitro.56 They did block α-thrombin–induced calcium mobilization and γ-thrombin–induced platelet aggregation in plasma in vitro.56 The minimal form of domain 4 that inhibited α-thrombin–induced platelet activation was the peptide RPPGF (Fig 5).56 RPPGF is the major angiotensin converting enzyme breakdown product of bradykinin in plasma with a metabolic degradation rate of 4.2 hours.242,243 The mechanism by which RPPGF and related domain 4 peptides inhibit α-thrombin–induced platelet activation is unique. RPPGF does not block the thrombin receptor peptide, SFLLRN, from inducing platelet activation.56 Domain 4 peptides prevent α-thrombin from cleaving the cloned thrombin receptor to initiate the activation process. This result means that domain 4 peptides actually prevented α-thrombin from cleaving the cloned thrombin receptor after arginine41 , a critical step in α-thrombin activation of cells through this receptor.56 When a peptide was prepared that spanned the α-thrombin cleavage site on the cloned thrombin receptor (NATLDPRSFLLR), RPPGF and HK actually prevented α-thrombin from cleaving this peptide between the arginine and the serine.56 RPPGF specifically interfered with thrombin's ability to cleave the cloned thrombin receptor to activate platelets without interfering with its procoagulant activity. These combined data indicate that domain 4 peptides and this same sequence in kininogens are selective, proteolytic inhibitors of α-thrombin–induced platelet activation by being directed to α-thrombin's substrate, the cloned thrombin receptor. Compounds based on the RPPGF sequence could represent a new class of thrombin inhibitors that achieve selectivity by being directed to the substrates of thrombin, rather than the enzyme itself.

Participation in Fibrinolysis

In addition to these unique mechanisms of α-thrombin inhibition, contact proteins participate in cellular fibrinolysis. From the time of recognition of HK deficiency, this protein has been ascribed to have a role in the fibrinolytic process, although the specific, physiologic mechanism has not been known.2,4 It has been known for more than 35 years that contact activation can increase total plasma fibrinolysis.244 Kallikrein, factor XIIa, and factor XIa cleave plasminogen directly, albeit much less efficiently than tPA or uPA.245-248 However, bradykinin has been characterized as a potent and selective in vivo inducer of tissue-type plasminogen activator release from endothelial cells in rabbits and humans.208,209 Plasma kallikrein also has been characterized to be a kinetically favorable activator of single-chain urokinase in vitro.116 More recent studies suggested single-chain urokinase activation by kallikrein can best occur on the platelet and endothelial cell surface.85,193,249 

These studies prompted us to examine the relationship of prekallikrein assembly on endothelial cells and how it may participate in single-chain urokinase activation (Table 2).86 When prekallikrein binds to HK on endothelial cells, the zymogen becomes activated to kallikrein, as indicated by elaboration of amidolytic activity, changes in the structure of prekallikrein to kallikrein on gel electrophoresis, and cleavage of HK.86 Prekallikrein activation occurs independently of any activated forms of factor XII. The prekallikrein-activating enzyme(s) is not a serine protease, but a membrane-associated or matrix-associated thiolprotease.86 Prekallikrein activation over endothelial cells is kinetically similar to prekallikrein activation by factor XII on an artificial surface. These data show for the first time that contact protein assembly on endothelial cells results in prekallikrein activation in the absence of factor XII and an artificial surface.86 This assembly of contact proteins allows for a physiologic pathway for this system to be activated. The degree of prekallikrein activation is regulated by HK. Increasing HK concentrations upregulates the enzyme that activates cell-bound prekallikrein. Thus, HK regulates prekallikrein activation, which, in turn, liberates more bradykinin from cell-bound HK and removes HK from the surface to slow prekallikrein activation.86 In support of this mechanism, we have recently shown that peptides derived from D6 of HK can downregulate plasmin formation by interfering with prekallikrein binding to HK on the endothelial cell surface.250 Also, increased bradykinin increases kininogen binding, which decreases soluble kallikrein from cleaving HK to liberate more bradykinin.35 Thus, there is a closely regulated pathway of prekallikrein activation and bradykinin liberation.

The prekallikrein activation pathway on endothelial cells participates in two pathways for fibrinolysis. First, kallikrein cleaves HK to liberate bradykinin, which is the most potent and specific stimulator of endothelial cell tissue-type plasminogen activator liberation.208,209 Second, kallikrein induces kinetically favorable conversion of single-chain urokinase into two-chain urokinase in an environment in which there is constitutive molar excess secretion of endothelial cell plasminogen activator inhibitor-1.86,250 Formation of two-chain urokinase results in a 4.3-fold increase in plasminogen activation. This system for plasminogen activation occurs in an environment in which there is no contribution by factor XIIa. This mechanism for single-chain urokinase activation is a pathway for cellular fibrinolysis that is either independent of or conjoined with single-chain urokinase activation associated with its binding to its receptor.251 The possible binding of HK (and, thus, kallikrein) to domain 2/3 of the urokinase receptor185 on the same molecule as prourokinase, which binds to domain 1 of the receptor, may allow for a very efficient cleavage of the latter by kallikrein. In addition, HK can compete with vitronectin, which also binds to domain 2/3 of the urokinase receptor, and displace vitronectin and its associated molecule, plasminogen activator inhibitor-1, thus enhancing fibrinolysis.

Antiadhesive Properties

HK has been postulated to be an antiadhesive protein. This property has been observed under three different situations. First, cleaved, kinin-free kininogen (HKa) can compete for deposition with adhesive proteins on artificial negatively charged surfaces such as those that occur on biomaterials. Second, HKa can compete with adhesive proteins for binding to cells. Third, HK on surfaces or in solution can prevent cells from attaching to protein-covered surfaces.

Vroman and Adams252 found that fibrinogen can be detected immunochemically on a negatively charged surface within seconds after normal human plasma contacts the surface, but, within minutes, is no longer detectable. We have shown that this phenomenon is due to the displacement of fibrinogen by HK after surface-dependent autoactivation of factor XII.253,254 Factor XIIa, both directly and indirectly (through the formation of kallikrein), generates HKa from HK. HKa (but not HK or LK) displaces fibrinogen from the surface.62 Therefore, the Vroman effect is due to the time- and surface-dependent generation of HKa, via contact activation of plasma, which results in the physical displacement of adherent fibrinogen from the surface.254 Extensive proteolysis results in HKi,62 which does not displace fibrinogen.254 

We have also described a similar effect on blood cells, as HK and/or HKa can displace 125I-fibrinogen from both neutrophils and platelets.175 Asakura et al33 extended these results by showing that HKa, but not HK, HKi, or LK, inhibited the adhesion and spreading of human osteosarcoma cells to vitronectin-coated polystyrene plates. HKa inhibited the attachment of platelets and monocytes to extracellular matrix proteins, and the spread of bovine aortic endothelial cells on both fibrinogen and vitronectin.33 The inhibition by HK of cell attachment to vitronectin may be explained by their competition for occupancy of domain 2/3 on the urokinase receptor.185 Results from our laboratory also indicated that neutrophils in a flow system at a shear rate of 20 s−1 adhere to a fibrinogen-coated surface linearly. In contrast, the rate of adherence to the same surface coated with HK is at least five times slower.255 The possibility of passivating surfaces with kininogen or its peptides may provide a new approach to biocompatibility. Alternatively, ligands derived from HK could, by binding to neutrophils, prevent their adhesion to surfaces or other cells such as endothelial cells.

Hereditary Angioedema (HAE)

HAE is a congenital condition associated with a deficiency or defect in C1 inhibitor (Table 3).256,257 Acute attacks of HAE have been well-documented to be associated with contact system activation.14,258-260 Characteristically, in acute attacks of HAE, there is reduced plasma prekallikrein activity with normal plasma prekallikrein antigen and reduced HK activity and antigen.14,259,261 Contact activation arises due to the absence of protease inhibition as a result of lowered C1 inhibitor levels. Bradykinin liberation is believed to be a major mediator of the edema seen in that condition.262 The phenomena of cold activation of factor VII is result of cold inactivation of C1 inhibitor and factor XII activation in a tube with resultant factor VII activation.263-265 Lowering temperatures to less than 37°C decreases the reactivity of C1 inhibitor for its enzymes.266 

Table 3.

Diseases States and Conditions Associated With Contact System Activation

Disease3-150PKHKC1 INHXI (Act.)XII (Act.)α2M-Kal Complexes (Ant.)C1 INH-Kal Complexes (Ant.)
Act.Ant.Act.Ant.Act.Ant.
HAE14,259  —   —   —  
Sepsis270,276   —   —   —   —   —   —  
Typhoid fever272   —   —   —   —   —   —  
ARDS269   —   —  
RMSF274   —   —   —   —  
Low-dose endotoxin277   —   —   —   —   —  
CPB281-283  —   —   —   —   —   —   —   —   —  
Disease3-150PKHKC1 INHXI (Act.)XII (Act.)α2M-Kal Complexes (Ant.)C1 INH-Kal Complexes (Ant.)
Act.Ant.Act.Ant.Act.Ant.
HAE14,259  —   —   —  
Sepsis270,276   —   —   —   —   —   —  
Typhoid fever272   —   —   —   —   —   —  
ARDS269   —   —  
RMSF274   —   —   —   —  
Low-dose endotoxin277   —   —   —   —   —  
CPB281-283  —   —   —   —   —   —   —   —   —  

Abbreviations: D, decreased; U, unaffected; I, increased.

F3-150

The following diseases were investigated: HAE, hereditary angiodema; ARDS, adult respiratory distress syndrome; RMSF, Rocky Mountain spotted fever; CPB, cardiopulmonary bypass. The numbers after the disease category are references.

Sepsis

Contact system activation has been postulated to be one of the mediators of systemic inflammatory response syndrome (SIRS).267 Contact activation of factor XII and prekallikrein in sepsis result in cleavages that activate them to enzymes that rapidly react with C1-inhibitor to form factor XIIa-C1-INH and kallikrein-C1-INH complexes (Table 3).268 The result is depletion of functional prekallikrein and factor XII with persistence of normal levels of the corresponding antigens. Functional C1-inhibitor also declines, but its antigen remains constant or may even increase, suggesting that it behaves as a weak acute-phase reactant. As functional C1-INH decreases, α2M becomes a more important inhibitor of kallikrein and α2M-Kal complexes form.14 The HK coagulant activity and antigen decrease in parallel.269 Paradoxically, for unknown reasons, functional factor XI may increase.269 

Investigations of patients with gram-negative sepsis showed that functional factor XII, prekallikrein, and C1-INH are decreased in patients with hypotensive septicemia.270 Patients with disseminated intravascular coagulation (DIC) due to septicemia or viremia had decreased functional factor XII, prekallikrein, and C1-INH, but individuals with DIC secondary to neoplasia had no significant changes in the kallikrein-kinin system.270 In patients with postoperative septicemia, decreased prekallikrein activity and elevated bradykinin were associated with positive blood cultures and hypotension.271 In an experimental infection of humans with typhoid fever, all patients with typhoid fever showed a decrease in functional prekallikrein and C1-INH, but the corresponding antigens remained unaffected.272 In the adult respiratory distress syndrome (ARDS), effected patients had reduced plasma levels of factor XII and prekallikrein.269,273 HK and C1-INH activity were also decreased, but there were increased levels of C1-INH antigen. Decreased levels of prekallikrein also have been documented in patients with septicemia due to viruses, fungi, or Rickettsia. Patients with Rocky Mountain Spotted Fever have decreased prekallikrein levels but increased kallikrein-C1-inhibitor complexes.274 Because, kallikrein-C1-inhibitor complexes are cleared rapidly in most cases of septic shock,275 we developed a sandwich enzyme-linked immunosorbent assay for α2M-Kal complexes and found that, in septicemic hypotension, but not in septicemia alone, α2M-Kal complexes were elevated.276 

None of the studies noted above indicated whether activation of the contact system is an early event or whether it is related to complications of sepsis such as hypotension and multiple organ failure. To address this question, normal human volunteers received a low dose of E coli endotoxin (0.4 ng/kg body weight). These individuals developed a flu-like illness associated with a hyperdynamic cardiovascular state lasting 24 hours.277 Functional prekallikrein levels were significantly lower in the endotoxin group as compared with controls at 2 hours after infusion and remained low throughout the rest of the experimental protocol at 5 and 24 hours. The concentration of α2M-Kal complexes was significantly elevated fourfold in the endotoxin-treated group by 3 hours and fivefold by 5 hours, with a decrease to normal in the circulating levels of complexes by 24 hours. Thus, a low dose of endotoxin can induce a prolonged state of contact activation.

To prove that contact activation is related to either shock or DIC, animal studies were performed (Table 4). In an established experimental baboon model of bacteremia, two concentrations of E coli were used to produce lethal and nonlethal hypotension. The lethal group developed irreversible hypotension, which significantly correlated with both the decline in functional levels of HK and an increase in α2M-Kal complexes.278 The nonlethal group experienced reversible hypotension, a less striking decline in HK, and only a slight elevation in α2M-Kal.278 Irreversible hypotension correlated with activation of the contact system. Further investigations were performed to address the causality of contact activation in shock and hypotension. An MoAb to human factor XII that is able in vitro to inhibit factor XII coagulant activity in baboon plasma by 60% and slow kininogen cleavage in dextran sulfate-activated baboon plasma was infused into the lethal baboon group 30 minutes before the E coli.279 Although the decline of factor V, fibrinogen, and platelets were similar in both groups and prekallikrein values were normal, there was a marked decline in HK in the untreated group, reaching 40% of the baseline levels by 300 minutes. In the group treated with the MoAb to factor XII, the HK remained stable and was significantly higher (110% of baseline) at 360 minutes. Furthermore, in the untreated group, there was a progressive increase of α2M-Kal complexes, which was highly significant and was completely blocked by the MoAb in the treated group. A significant decline of mean systemic arterial pressure was observed in both groups of animals between 60 and 120 minutes. A Kaplan-Meier plot showed that treated animals survived significantly longer than untreated animals. Inhibition of contact system activation with an MoAb to factor XII modulated the hypotension.279 

Table 4.

Experimental Diseases States for Which Contact System Activation Is Pathogenetic

Disease4-150PKHKC1 INHXI (Act.)XII (Act.)α2M-Kal Complexes (Ant.)C1 INH-Kal Complexes (Ant.)
Act.Ant.Act.Ant.Act.Ant.
Baboon sepsis278,279   —   —   —   —   —   —  
Rat arthritis284,286   —   —   —   —   —   —   —  
Rat enterocolitis286,287   —   —   —   —   —   —   —  
Disease4-150PKHKC1 INHXI (Act.)XII (Act.)α2M-Kal Complexes (Ant.)C1 INH-Kal Complexes (Ant.)
Act.Ant.Act.Ant.Act.Ant.
Baboon sepsis278,279   —   —   —   —   —   —  
Rat arthritis284,286   —   —   —   —   —   —   —  
Rat enterocolitis286,287   —   —   —   —   —   —   —  

In these conditions, a role for contact activation in the pathogenesis of these disorders is shown by the finding that specific inhibitors of the contact system blocked the progression of the disease.

Abbreviations: D, decreased; I, increased.

F4-150

The numbers after the disease category are references.

Cardiopulmonary Bypass

Clinical cardiopulmonary bypass (CPB) is performed on more than 350,000 Americans each year (Table 3). During CPB, there is extensive contact between blood anticoagulated with heparin and the synthetic surfaces of the extracorporeal circuit. Blood cell interactions and plasma protein alterations prolong the bleeding time, increase postoperative blood loss, and trigger a chemical and cellular whole body inflammatory response. Extracorporeal circulation has been associated with both qualitative and quantitative alterations of platelets, neutrophils and complement and contact systems. Heparin, which markedly accelerates inactivation of FXa and thrombin by antithrombin III, exhibits minimal enhancement of the inactivation of FXIIa and FXIa in CPB.280 In simulated CPB, there is a significant increase in kallikrein-C1-INH complex formation.281 The simultaneous formation of C1-C1-INH complexes suggested that factor XII activation occurred, which, in turn, activated both kallikrein and C1, thus triggering both the contact and classical complement pathways.281 Further studies showed that aprotinin, an inhibitor of both plasmin and plasma kallikrein, reduced blood loss after cardiac operations and decreased the elevated postoperative bleeding time. In a simulated extracorporeal bypass model, in which no plasmin is found, aprotinin decreased both kallikrein-C1-inhibitor and C1-C1-inhibitor complexes, resulting in a marked inhibition of the release of neutrophil elastase.282 Similar results were obtained with specific kallikrein inhibitors, Bz-Pro-Phe-boroArg-OH, Arg15-aprotinin, and Ala357-Arg358-α1 -protease inhibitor.283 

Experimental Arthritis in Genetically Susceptible Rats

The role of the kallikrein-kinin system in inflammatory arthritis was investigated by a model of acute and chronic relapsing arthritis induced by intraperitoneal injection of proteoglycan-polysaccharide from group A streptococci (PG-APS) into rats (Table 4).284 The mean joint diameter peaked at a maximum value of 8 at day 3, indicating an acute arthritis. After a decrease in the volume of the joint on days 9 through 12, the joint diameter spontaneously increased beginning at day 15 and then progressed with waxing and waning of individual joints, indicating reactivation leading to chronic synovitis and joint erosion. An increase in the acute-phase protein, T-kininogen, splenic enlargement, and the development of the anemia of chronic disease were consistently associated with the arthropathy. HK in rat plasma decreased on days 1, 5, and 15, but not at 30 minutes, day 23, or day 45. There is a striking inverse correlation between HK concentration and joint enlargment on day 5, with r = .85. Prekallikrein levels were significantly lower in PG-APS–injected animals compared with controls. Prekallikrein levels decreased as early as 30 minutes after injection, and the levels remained low throughout the experimental protocol. Further experiments showed that, when the rats were injected with PG-APS and received a specific, potent oral plasma kallikrein inhibitor, P8720 or Bz-Pro-Phe-boroArg-OH (Ki = 0.15 nmol/L, Kassoc = 1.6 × 106 mol/Ls−1), there was a significant decrease (61%) in joint swelling at 49 hours, with a disappearance of most of the dense infiltration of neutrophils and mononuclear cells.285 Furthermore, there was no decrease in plasma HK. Lastly, the anemia, the increase of TK, and the splenic weight increase were largely inhibited. These data indicated that contact system activation mediates the arthritis and that its inhibition ameliorates all the manifestations of this disorder.

Acute and Chronic Enterocolitis in Genetically Susceptible Rats

Further investigations examined the role of the contact system in inflammatory bowel disease using a model of acute and chronic enterocolitis induced by subserosal injection of PG-APS into the wall of the distal ileum and cecum (Table 4).286 Acute intestinal inflammation in the Lewis rat and the Buffalo rat are characterized by edema, hemorrhage, thickening of the bowel wall, and mesentery and adhesions. However, genetically susceptible Lewis rats, but not resistant Buffalo rats, spontaneously develop chronic enterocolitis with dense adhesions, thickening of intestinal wall, serosal nodules, enlarged mesenteric lymph nodes, histological changes consisting of mononuclear cell infiltration and crypt abscesses, and a markedly elevated intestinal myeloperoxidase that persists for at least 16 weeks. Furthermore, a marked disparity existed in the incidence of extraintestinal manifestations between Lewis and Buffalo rats. Arthritis and hepatic granulomas occurred in 73% of Lewis rats examined 14 days or more after PG-APS injection; however, only 4% of Buffalo rats developed hepatic granulomas, and arthritis was not evident in Buffalo rats. T-kininogen, the major acute-phase protein in the rat, increased in the Lewis rat but not in the Buffalo rat.

PK levels were significantly lower in PG-APS–treated Lewis rats compared with controls or Buffalo rats during both acute and chronic phases of inflammation. HK levels were significantly decreased in PG-APS–treated Lewis rats on days 5 and 42 from its respective control group. Buffalo rats injected with PG-APS had stable plasma HK concentrations at all time points. Treatment with the specific, oral plasma kallikrein inhibitor, Bz-Pro-Phe-boroArg-OH, in the acute phase of enterocolitis287 in the Lewis rat decreased the increase of joint diameter, the gross gut score, and intestinal myeloperoxidase activity and prevented the decrease of factor XI and HK. The kallikrein inhibitor could block bradykinin release and, thus, pain, swelling, and vasodilation as well as neutrophil activation. Moreover, because factor XII can stimulate IL-1 expression, IL-1 induces IL-6 expression, and an MoAb to factor XIIa blocks IL-6 release, one can surmise that contact activation is involved in the acute and chronic phases of the enterocolitis.204,288 Demonstration of a pathogenetic role of the kallikrein-kinin system in experimental enterocolitis, arthritis, and related systemic inflammation suggest a similar role in idiopathic human intestinal and joint inflammation and raise the possibility that selective kallikrein inhibitors may be useful in disorders such as Crohn's disease and rheumatoid arthritis.

Thrombosis Risk Factor

Independent of the multiple mechanisms by which kininogens are selective antithrombins that modulate α-thrombin's activation of platelets and endothelial cells in vitro and the proposed physiologic mechanism for cellular fibrinolysis due to assembly of HK and PK on endothelium, clinical observation suggests that deficiencies of these proteins may be additive risk factors for thrombosis. John Hageman (Hageman factor) and Mayme Williams (Williams trait) both died of pulmonary emboli. Although both of these patients had other reasons for pulmonary emboli, their contact protein deficiencies may have contributed an additional risk factor. Certainly, their deficiencies did not protect them from thrombosis. Numerous other clinical studies also suggest that contact protein deficiencies may be associated with impaired contact factor-dependent fibrinolysis. This result may contribute to an increased incidence of thrombosis in patients with congenital factor XII deficiency,289-293 an increased incidence of factor XII deficiency in patients with venous thrombosis, and acquired thrombotic disorders such as myocardial infarction294 and re-thrombosis of coronary arteries after thrombolytic therapy.295 Although these studies are interesting, contact protein deficiencies are relatively rare occurrences. It will require careful prospective investigations with age- and sex-matched controls to determine whether these factors contribute to the ever enlarging list of inherited risk factors for thrombosis.

Supported in part by National Institutes of Health Grants No. HL54894, DK43735, and HL47186 to R.W.C. and HL35553, HL52799, HL55907, and HL56415 to A.H.S.

Address reprint requests to Alvin H. Schmaier, MD, University of Michigan 5301 MSRBIII, 1150 W Medical Center Dr, Ann Arbor, MI 48109-0640.

1
Roche e Silva
M
Beraldo
WT
Rosenfeld
G
Bradykinin, a hypotensive and smooth muscle stimulating factor released from plasma globulin by snake venoms and by trypsin.
Am J Physiol
150
1949
261
2
Colman
RW
Bagdasarian
A
Talamo
RC
Scott
CF
Seavey
M
Guimaraes
JA
Pierce
JV
Kaplan
AP
Williams trait. Human kininogen deficiency with diminished levels of plasminogen proactivator and prekallikrein associated with abnormalities of the Hageman factor-dependent pathways.
J Clin Invest
56
1975
1650
3
Wuepper
KD
Miller
DR
Lacombe
MJ
Flaujeac trait. Deficiency of human plasma kininogen.
J Clin Invest
56
1975
1663
4
Saito
H
Ratnoff
OD
Waldmann
R
Abraham
JP
Fitzgerald trait: Deficiency of a hitherto unrecognized agent, Fitzgerald factor, participating in surface mediated reactions of clotting, fibrinolysis, generation of kinins, and the property of diluted plasma enhancing vascular permeability (PF/DIL).
J Clin Invest
55
1975
1082
5
Takagaki
Y
Kitamura
N
Nakanishi
S
Cloning and sequence analysis of cDNAs for human high molecular weight and low molecular weight prekininogens. Primary structures of two human prekininogens.
J Biol Chem
260
1985
8601
6
Kitamura
N
Kitagawa
H
Fukushima
D
Takagaki
Y
Miyata
T
Nakanishi
S
Structural organization of the human kininogen gene and a model for its evolution.
J Biol Chem
260
1985
8610
7
Fong
D
Smith
DI
Hsieh
WT
The human kininogen gene (KNG) mapped to chromosome 3q26-qter by analysis of somatic cell hybrids using the polymerase chain reaction.
Hum Genet
87
1991
189
8
Cheung
PP
Cannizzaro
LA
Colman
RW
Chromosomal mapping of human kininogen gene (KNG) to 3q26→qter.
Cytogenet Cell Genet
59
1992
24
9
Rizzu
P
Baldini
A
Three members of the human cystatin gene superfamily, AHSG, HRG, and KNG, map within one megabase of genomic DNA at 3q27.
Cytogenet Cell Genet
70
1995
26
10
Kakizuka
A
Ingi
T
Murai
T
Nakanishi
S
A set of U1 snRNA-complementary sequences involved in governing alternative RNA splicing of the kininogen genes.
J Biol Chem
265
1990
10102
11
Cheung
PP
Kunapuli
SP
Scott
CF
Wachtfogel
YT
Colman
RW
Genetic basis of total kininogen deficiency in Williams' trait.
J Biol Chem
268
1993
23361
12
Hayashi
I
Hoshiko
S
Makabe
O
Oh-ishi S
A point mutation of alanine 163 to threonine is responsible for the defective secretion of high molecular weight kininogen by the liver of brown Norway Katholiek rats.
J Biol Chem
268
1993
17219
13
Chen
LM
Chung
P
Chao
S
Chao
L
Chao
J
Differential regulation of kininogen gene expression by estrogen and progesterone in vivo.
Biochim Biophys Acta
1131
1992
145
14
Chhibber
G
Cohen
A
Lane
S
Farber
A
Meloni
FJ
Schmaier
AH
Immunoblotting of plasma in a pregnant patient with hereditary angioedema.
J Lab Clin Med
115
1990
112
15
Takano
M
Yokoyama
K
Yayama
K
Okamoto
H
Murine fibroblasts synthesize and secrete kininogen in response to cyclic-AMP, prostaglandin E2 and tumor necrosis factor.
Biochim Biophys Acta
1265
1995
189
16
Scott
CF
Colman
RW
Sensitive antigenic determinations of high molecular weight kininogen performed by covalent coupling of capture antibody.
J Lab Clin Med
119
1992
77
17
Jacobsen
S
Kriz
M
Some data on two purified kininogens from human plasma.
Br J Pharmacol
29
1967
25
18
Schmaier
AH
Bradford
H
Silver
LD
Farber
A
Scott
CF
Schutsky
D
Colman
RW
High molecular weight kininogen is an inhibitor of platelet calpain.
J Clin Invest
77
1986
1565
19
Schmaier
AH
Zuckerberg
A
Silverman
C
Kuchibhotla
J
Tuszynski
GP
Colman
RW
High-molecular weight kininogen. A secreted platelet protein.
J Clin Invest
71
1983
1477
20
Schmaier
AH
Kuo
A
Lundberg
D
Murray
S
Cines
DB
The expression of high molecular weight kininogen on human umbilical vein endothelial cells.
J Biol Chem
263
1988
16327
21
Gustafson
EJ
Schmaier
AH
Wachtfogel
YT
Kaufman
N
Kucich
U
Colman
RW
Human neutrophils contain and bind high molecular weight kininogen.
J Clin Invest
84
1989
28
22
Proud
D
Perkins
M
Pierce
JV
Yates
KN
Highet
PF
Herring
PL
Mangkornkanok/Mark M
Bahu
R
Carone
F
Pisano
JJ
Characterization and localization of human renal kininogen.
J Biol Chem
256
1981
10634
23
Hallbach
J
Adams
G
Wirthensohn
G
Guder
WG
Quantification of kininogen in human renal medulla.
Biol Chem Hoppe Seyler
368
1987
1151
24
Yamamoto
T
Tsuruta
J
Kambara
T
Interstitial-tissue localization of high-molecular-weight kininogen in guinea-pig skin.
Biochim Biophys Acta
916
1987
332
25
Ohkubo
I
Kurachi
K
Takasawa
T
Shiokawa
H
Sasaki
M
Isolation of a human cDNA for alpha 2-thiol proteinase inhibitor and its identity with low molecular weight kininogen.
Biochemistry
23
1984
5691
26
Weisel
JW
Nagaswami
C
Woodhead
JL
DeLa
Cadena RA
Page
JD
Colman
RW
The shape of high molecular weight kininogen: Organization into structural domains, changes with activation, and interactions with prekallikrein, as determined by electron microscopy.
J Biol Chem
269
1994
10100
27
Salvesen
G
Parkes
C
Abrahamson
M
Grubb
A
Barrett
AJ
Human low-Mr kininogen contains three copies of a cystatin sequence that are divergent in structure and in inhibitory activity for cysteine proteinases.
Biochem J
234
1986
429
28
Schmaier
AH
Schutsky
D
Farber
A
Silver
LD
Bradford
HN
Colman
RW
Determination of the bifunctional properties of high molecular weight kininogen by studies with monoclonal antibodies directed to each of its chains.
J Biol Chem
262
1987
1405
29
Vogel
R
Assfalg-Machleidt
I
Esterl
A
Machleidt
W
Muller-Esterl
W
Proteinase-sensitive regions in the heavy chain of low molecular weight kininogen map to the inter-domain junctions.
J Biol Chem
263
1988
12661
30
Bradford
HN
Jameson
BA
Adam
AA
Wassell
RP
Colman
RW
Contiguous binding and inhibitory sites on kininogens required for the inhibition of platelet calpain.
J Biol Chem
268
1993
26546
31
Hasan
AA
Cines
DB
Ngaiza
JR
Jaffe
EA
Schmaier
AH
High-molecular-weight kininogen is exclusively membrane bound on endothelial cells to influence activation of vascular endothelium.
Blood
85
1995
3134
32
Hasan
AA
Zhang
J
Samuel
M
Schmaier
AH
Conformational changes in low molecular weight kininogen alters its ability to bind to endothelial cells.
Thromb Haemost
74
1995
1088
33
Asakura
S
Hurley
RW
Skorstengaard
K
Ohkubo
I
Mosher
DF
Inhibition of cell adhesion by high molecular weight kininogen.
J Cell Biol
116
1992
465
34
Ishiguro
H
Higashiyama
S
Ohkubo
I
Sasaki
M
Heavy chain of human high molecular weight and low molecular weight kininogen binds calcium ion.
Biochemistry
26
1987
7021
35
Zini
JM
Schmaier
AH
Cines
DB
Bradykinin regulates the expression of kininogen binding sites on endothelial cells.
Blood
81
1993
2936
36
Gustafson
EJ
Schutsky
D
Knight
L
Schmaier
AH
High molecular weight kininogen binds to unstimulated platelets.
J Clin Invest
78
1986
310
37
Greengard
JS
Heeb
MJ
Ersdal
E
Walsh
PN
Griffin
JH
Binding of coagulation factor XI to washed human platelets.
Biochemistry
25
1986
3884
38
van Iwaarden
F
deGroot
PG
Bouma
BN
The binding of high molecular weight kininogen to cultured human endothelial cells.
J Biol Chem
263
1988
4698
39
Croxatto
HR
Boric
MP
Roblero
J
Albertini
R
Silva
R
Digestive process and regulation of renal excretory function. Pepsanurin and prokinins inhibitors of diuresis mediated by atrial natriuretic peptide.
Rev Med Chile
122
1995
1162
40
Bradford
HN
Schmaier
AH
Colman
RW
Kinetics of inhibition of platelet calpain II by human kininogens.
Biochem J
270
1990
83
41
Auerswald
EA
Rossler
D
Mentele
R
Assfalg-Machleidt
I
Cloning, expression and characterization of human kininogen domain 3.
FEBS Lett
321
1993
93
42
Ylinenjarvi
K
Prasthofer
TW
Martin
NC
Bjork
I
Interaction of cysteine proteinases with recombinant kininogen domain 2, expressed in Escherichia coli.
FEBS Lett
357
1995
309
43
Bano
B
Kunapuli
SP
Bradford
HN
Colman
RW
Structural requirements for cathepsin B and cathepsin H inhibition by kininogens.
J Protein Chem
15
1996
519
44
Scott
CF
Whitaker
EJ
Hammond
BF
Colman
RW
Purification and characterization of a potent 70-kDa thiol lysyl-proteinase (Lys-gingivain) from Porphyromonas gingivalis that cleaves kininogens and fibrinogen.
J Biol Chem
268
1993
7935
45
Schmaier
AH
Smith
PM
Purdon
AD
White
JG
Colman
RW
High molecular weight kininogen: Localization in the unstimulated and activated platelet and activation by a platelet calpain(s).
Blood
67
1986
119
46
Schmaier
AH
Bradford
HN
Lundberg
D
Farber
A
Colman
RW
Membrane expression of platelet calpain.
Blood
75
1990
1273
47
Saido
T
Suzuki
H
Yamazaki
H
Tanoue
K
Suzuki
K
In situ capture of m-calpain activation of platelets.
J Biol Chem
268
1993
7422
48
Meloni
FJ
Schmaier
AH
Low molecular weight kininogen binds to platelets to modulate thrombin-induced platelet activation.
J Biol Chem
266
1991
6786
49
Reddigari
SR
Kuna
P
Miragliotta
G
Shibayama
Y
Nishikawa
K
Kaplan
AP
Human high molecular weight kininogen binds to human umbilical vein endothelial cells via its heavy and light chains.
Blood
81
1993
1306
50
Jiang
YP
Muller
Esterl W
Schmaier
AH
Domain 3 of kininogens contains a cell-binding site and a site that modifies thrombin activation of platelets.
J Biol Chem
267
1992
3712
51
Wachtfogel
YT
DeLa
Cadena RA
Kunapuli
SP
Rick
L
Miller
M
Schultze
RL
Altieri
DC
Edgington
TS
Colman
RW
High molecular weight kininogen binds to Mac-1 on neutrophils by its heavy chain (domain 3) and its light chain (domain 5).
J Biol Chem
269
1994
19307
52
Bode
W
Engh
R
Musil
D
Thiele
U
Huber
R
Karshikov
A
Brzin
J
Kos
J
Turk
V
The 2.0 A X-ray crystal structure of chicken egg white cystatin and its possible mode of interaction with cysteine proteinases.
EMBO J
7
1988
2593
53
Herwald
H
Hasan
AAK
Godovac-Zimmermann
J
Schmaier
AH
Muller-Esterl
W
Identification of an endothelial cell binding site on kininogen domain D3.
J Biol Chem
270
1995
14634
54
DeLa
Cadena RA
Wyshock
EG
Kunapuli
SP
Schultze
RL
Miller
M
Walz
DA
Colman
RW
Platelet thrombospondin interactions with human high and low molecular weight kininogens.
Thromb Haemost
72
1994
125
55
Puri
RN
Zhou
F
Hu
CJ
Colman
RF
Colman
RW
High molecular weight kininogen inhibits thrombin-induced platelet aggregation and cleavage of aggregin by inhibiting binding of thrombin to platelets.
Blood
77
1991
500
56
Hasan
AAK
Amenta
S
Schmaier
AH
Bradykinin and its metabolite, Arg-Pro-Pro-Gly-Phe, are selective inhibitors of α-thrombin-induced platelet activation.
Circulation
94
1996
517
57
Kunapuli
SP
Bradford
HN
Jameson
BA
DeLa
Cadena RA
Rick
L
Wassell
RP
Colman
RW
Thrombin-induced platelet aggregation is inhibited by the heptapeptide Leu271-Ala277 domain 3 in the heavy chain of high molecular weight kininogen.
J Biol Chem
271
1996
11228
58
Mori
K
Nagasawa
S
Studies on human high molecular weight (HMW) kininogen. II. Structural change of HMW kininogen by the action of human plasma kallikrein.
J Biochem
89
1981
1465
59
Schmaier
AH
Farber
A
Schein
R
Sprung
C
Structural changes of plasma high molecular weight kininogen after in vitro activation and in sepsis.
J Lab Clin Med
112
1988
182
60
Tayeh
MA
Olson
ST
Shore
JD
Surface-induced alterations in the kinetic pathway for cleavage of human high molecular weight kininogen by plasma kallikrein.
J Biol Chem
269
1994
16318
61
Wiggins
RC
Kinin release from high molecular weight kininogen by the action of Hageman factor in the absence of kallikrein.
J Biol Chem
258
1983
8963
62
Scott
CF
Silver
LD
Purdon
AD
Colman
RW
Cleavage of human high molecular weight kininogen by factor XIa in vitro. Effect on structure and function.
J Biol Chem
260
1985
10856
63
Sato
F
Nagasawa
S
Mechanism of kinin release from human low-molecular-mass-kininogen by the synergistic action of human plasma kallikrein and leukocyte elastase.
Biol Chem Hoppe Seyler
369
1988
1009
64
Kleniewski
J
Donaldson
VH
Granulocyte elastase cleaves human high molecular weight kininogen and destroys its clot-promoting activity.
J Exp Med
167
1988
1895
65
Hasan
AAK
Cines
DB
Zhang
J
Schmaier
AH
The carboxyl terminus of bradykinin and amino terminus of the light chain of kininogens comprise an endothelial cell binding domain.
J Biol Chem
269
1994
31822
66
Meloni
FJ
Gustafson
EJ
Schmaier
AH
High molecular weight kininogen binds to platelets by its heavy and light chains and when bound has altered susceptibility to kallikrein cleavage.
Blood
79
1992
1233
67
Hasan
AAK
Cines
DB
Herwald
H
Schmaier
AH
Muller-Esterl
W
Mapping the cell binding site on high molecular weight kininogen domain 5.
J Biol Chem
270
1995
19256
68
Khan M, Punia N, Majluf-Cruz A, Kunapuli SP, Cooper SL, DeLa Cadena RA, Colman RW: The binding sites on high molecular weight kininogen (HK) to activated human neutrophils are localized to K263-Q292, Q329-M357, and H493-K520. Blood 86:33a, 1995 (abstr, suppl 1)
69
Retzios
AD
Rosenfeld
R
Schiffman
S
Effects of chemical modifications on the surface- and protein-binding properties of the light chain of human high molecular weight kininogen.
J Biol Chem
262
1987
3074
70
Kunapuli
SP
DeLa
Cadena RA
Colman
RW
Deletion mutagenesis of high molecular weight kininogen light chain: Identification of two anionic surface binding subdomains.
J Biol Chem
268
1993
2486
71
Bjork
I
Olson
ST
Sheffer
RG
Shore
JD
Binding of heparin to human high molecular weight kininogen.
Biochemistry
28
1989
1213
72
DeLa
Cadena RA
Colman
RW
The sequence HGLGHGHEQQHGLGHGH in the light chain of high molecular weight kininogen serves as a primary structural feature for zinc-dependent binding to an anionic surface.
Protein Sci
1
1992
151
73
Ben
Nasr AB
Herwald
H
Muller-Esterl
W
Bjorck
L
Human kininogens interact with M protein, a bacterial surface protein and virulence determinant.
Biochem J
305
1995
173
74
Scott
CF
Silver
LD
Schapira
M
Colman
RW
Cleavage of human high molecular weight kininogen markedly enhances its coagulant activity. Evidence that this molecule exists as a procofactor.
J Clin Invest
73
1984
954
75
Tait
JF
Fujikawa
K
Identification of the binding site for plasma prekallikrein in human high molecular weight kininogen. A region from residues 185 to 224 of the kininogen light chain retains full binding activity.
J Biol Chem
261
1986
15396
76
Tait
JF
Fujikawa
K
Primary structure requirements for the binding of human high molecular weight kininogen to plasma prekallikrein and factor XI.
J Biol Chem
262
1987
11651
77
Vogel
R
Kaufmann
J
Chung
DW
Kellermann
J
Muller-Esterl
W
Mapping of the prekallikrein binding site of human H-kininogen by ligand screening of lambda gt11 expression libraries: Mimicking of the predicted binding site by anti-idiotypic antibodies.
J Biol Chem
265
1990
12494
78
Bock
PE
Shore
JD
Protein-protein interactions in contact activation of blood coagulation. Characterization of fluorescein-labeled human high molecular weight kininogen-light chain as a probe.
J Biol Chem
258
1983
15079
79
Bock
PE
Shore
JD
Tans
G
Griffin
JH
Protein-protein interactions in contact activation of blood coagulation. Binding of high molecular weight kininogen and the 5-(iodoacetamido) fluorescein-labeled kininogen light chain to prekallikrein, kallikrein, and the separated kallikrein heavy and light chains.
J Biol Chem
260
1985
12434
80
Scarsdale
JN
Harris
RB
Solution phase conformation studies of the prekallikrein binding domain of high molecular weight kininogen.
J Protein Chem
9
1990
647
81
You
JL
Scarsdale
JN
Harris
RB
Calorimetric and spectroscopic examination of the solution phase structures of prekallikrein binding domain peptides of high molecular weight kininogen.
J Protein Chem
10
1991
301
82
You
JL
Page
JD
Scarsdale
JN
Colman
RW
Harris
RB
Conformational analysis of synthetic peptides encompassing the factor XI and prekallikrein overlapping binding domains of high molecular weight kininogen.
Peptides
14
1993
867
83
Reddigari
S
Kaplan
AP
Monoclonal antibody to human high molecular weight kininogen recognizes its prekallikrein binding site and inhibits coagulant activity.
Blood
74
1989
695
84
Kaufmann
J
Haasemann
M
Modrow
S
Muller-Esterl
W
Structural dissection of the multidomain kininogens. Fine mapping of the target epitopes of antibodies interfering with their functional properties.
J Biol Chem
268
1993
9079
85
Lenich
C
Pannell
R
Gurewich
V
Assembly and activation of the intrinsic fibrinolytic pathway on the surface of human endothelial cells in culture.
Thromb Haemost
74
1995
698
86
Motta G, Rojkjaer R, Hasan AAK, Cines DB, Schmaier AH: High molecular weight kininogen regulates prekallikrein assembly and activation on endothelial cells: A novel mechanism for contact activation. Blood (in press)
87
Beaubien
G
Rosinski-Chupin
I
Mattei
MG
Mbikay
M
Chretien
M
Seidah
NG
Gene structure and chromosomal localization of plasma kallikrein.
Biochemistry
30
1991
1628
88
Asakai
R
Davie
EW
Chung
DW
Organization of the gene for human factor XI.
Biochemistry
26
1987
7221
89
Chung
DW
Fujikawa
K
McMullen
BA
DAvie
EW
Human plasma prekallikrein, a zymogen to a serine protease that contains four tandem repeats.
Biochemistry
25
1986
2410
90
McMullen
BA
Fujikawa
K
Davie
EW
Location of the disulfide bonds in human plasma prekallikrein: The presence of four novel apple domains in the amino-terminal portion of the molecule.
Biochemistry
30
1991
2050
91
McMullen
BA
Fugikawa
K
Davie
EW
Location of the disulfide bonds in human coagulation factor XI: The presence of a Tandem apple domain.
Biochemistry
30
1991
2056
92
Mandle
RJ Jr
Kaplan
AP
Hageman factor substrates. Human plasma prekallikrein: Mechanism of activation by Hageman factor and participation in Hageman factor-dependent fibrinolysis.
J Biol Chem
252
1977
6097
93
Scott
CF
Liu
CY
Colman
RW
Human plasma prekallikrein: A rapid high yield method for purification.
Eur J Biochem
100
1979
77
94
Fisher
CA
Schmaier
AH
Addonizio
VP
Colman
RW
Assay of prekallikrein in human plasma: Comparison of amidolytic, esterolytic, coagulation, and immunochemical assays.
Blood
59
1982
963
95
McConnell
DJ
Mason
B
The isolation of human plasma prekallikrein.
Br J Pharmacol
38
1970
490
96
Wuepper
KD
Cochrane
CG
Plasma prekallikrein: Isolation, characterization, and mechanism of action.
J Exp Med
135
1972
1
97
Burger
D
Schleuning
W-D
Schapira
M
Human plasma prekallikrein: Immunoaffinity purification and activation to α- and β-kallikrein.
J Biol Chem
261
1986
324
98
Colman
RW
Wachtfogel
YT
Kucich
U
Weinbaum
G
Hahn
S
Pixley
RA
Scott
CF
de Agostini
A
Burger
D
Schapira
M
Effect of cleavage of the heavy chain of human plasma kallikrein on its functional properties.
Blood
65
1985
311
99
Mandle
R Jr
Colman
RW
Kaplan
AP
Identification of prekallikrein and high molecular weight kininogen as a complex in human plasma.
Proc Natl Acad Sci USA
73
1976
4179
100
Page
JD
Colman
RW
Localization of distinct functional domains on prekallikrein for interaction with both high molecular weight kininogen and activated factor XII in a 28-kDa fragment (amino acids 141-371).
J Biol Chem
266
1991
8143
101
Page
JD
You
JL
Harris
RB
Colman
RW
Localization of the binding site on plasma kallikrein for high molecular weight kininogen to both apple 1 and apple 4 domains of the heavy chain.
Arch Biochem Biophys
314
1994
159
102
Hock J, Vogel R, Linke RP, Muller-Esterl W: High molecular weight kininogen-binding site of prekallikrein probed by monoclonal antibodies. J Biol Chem. 265:12005, 1990
103
Lin
Y
Shenoy
S
Harris
RB
Colman
RW
Direct evidence for multi-facial contacts between high molecular weight kininogen and plasma prekallikrein.
Biochemistry
35
1996
12945
104
Herwald
H
Renne
T
Meijers
JCM
Chung
D
Page
JD
Colman
RW
Muller-Esterl
W
Mapping of the discontinuous kininogen binding site of prekallikrein.
J Biol Chem
271
1996
13061
105
Gigli
I
Mason
JW
Colman
RW
Austen
KF
Interaction of plasma kallikrein with the C1 inhibitor.
J Immunol
104
1970
574
106
Van der Graff
F
Koedam
JA
Bouma
BA
Inactivation of Kallikrein in plasma.
J Clin Invest
71
1983
149
107
Schapira
M
Scott
CF
Colman
RW
Contribution of plasma protease inhibitors to the inactivation of kallikrein in plasma.
J Clin Invest
69
1982
462
108
Van der Graff
F
Koedam
JA
Griffin
JH
Bouma
BN
Interaction of human plasma, kallikrein and its light chain with C1 inhibitor.
Biochemistry
22
1983
4860
109
Schapira
M
Scott
CF
James
A
Silver
LD
Kueppers
F
James
HL
Colman
RW
High molecular weight kininogen or its light chain protects human plasma kallikrein from inactivation by plasma protease inhibitors.
Biochemistry
21
1982
567
110
Schapira
M
Scott
CF
Colman
RW
Protection of human plasma kallikrein from inactivation by C1 inhibitor and other protease inhibitors. The role of high molecular weight kininogen.
Biochemistry
20
1981
2738
111
Schmaier
AH
Gustafson
EG
Idell
S
Colman
RW
Plasma prekallikrein assay: Reversible inhibition of C1 inhibitor by chloroform and its use in measuring prekallikrein in different mammalian species.
J Lab Clin Med
104
1984
882
112
Lahiri
B
Bagdasarian
A
Mitchell
B
Rosenberg
RD
Colman
RW
Antithrombin-heparin cofactor: An inhibitor of plasma Kallikrein.
Arch Biochem Biophys
175
1976
737
113
Meijers
JCM
Kanters
DHAJ
Vlooswijk
RAA
Erp
HEV
Hessing
M
Bouma
BN
Inactivation of human plasma kallikrein and factor XIa by protein C inhibitor.
Biochemistry
27
1988
4231
114
Espana
F
Estelles
A
Griffin
JH
Aznar
J
Interaction of plasma kallikrein with protein C inhibitor in purified mixtures and in plasma.
Thromb Haemost
65
1991
46
115
Hauert
J
Nicoloso
G
Schleuning
WD
Bachmann
F
Schapira
M
Plasminogen activators in dextran sulfate-activated euglobulin functions: A molecular analysis of factor XII and prekallikrein-dependent fibrinolysis.
Blood
73
1989
994
116
Ichinose
A
Fujikawa
K
Suyama
T
The activation of prourokinase by plasma kallikrein and its inactivation by thrombin.
J Biol Chem
261
1986
3486
117
Citarella
F
Tripodi
M
Fantoni
A
Bernardi
F
Romeo
G
Rocchi
M
Assignment of human coagulation factor XII (fXII) to chromosome 5 by cDNA hybridization to DNA from somatic cell hybrids.
Hum Genet
80
1988
397
118
Rolye
NJ
Nigli
M
Cool
D
MacGilivray
RTA
Hamerton
JL
Structural gene encosing human factor XII is located at 5q33-qter.
Somat Cell Mol Genet
14
1988
217
119
Cool
DE
MacGillivray
RTA
Characterization of the human blood coagulation factor XII gene.
J Biol Chem
262
1987
13662
120
Cool
DE
Edgell
C-JS
Louie
GV
Zoller
MJ
Brayer
GD
MacGillivray
RTA
Characterization of human blood coagulation factor XII cDNA.
J Biol Chem
260
1985
13666
121
Que
BG
Davie
EW
Characterization of a cDNA coding for human factor XII (Hageman factor).
Biochemistry
25
1986
1525
122
Griffin
JH
Cochrane
CG
Human factor XII (Hageman factor).
Methods Enzymol
45
1976
56
123
Revak
SD
Cochrane
CC
Johnston
A
Hugli
T
Structural changes accompanying enzymatic activation of Hageman factor.
J Clin Invest
54
1974
619
124
Saito
H
Ratnoff
OD
Pensky
J
Radioimmunoassay of human Hageman factor (factor XII).
J Lab Clin Med
88
1976
506
125
Gordon
EM
Gallagher
CA
Johnson
TR
Blossey
BK
Ilan
J
Hepatocytes express blood coagulation factor XII (Hageman factor).
J Lab Clin Med
115
1990
463
126
Gordon
EM
Williams
SR
Frenchek
B
Mazur
CA
Speroff
L
Dose-dependent effects of postmenopausal estrogen and progestin on antithrombin III and factor XII.
J Lab Clin Med
111
1988
52
127
Mitropoulos
KA
Martin
JC
Burgess
Al
Esnouf
MP
Stirling
Y
Howorth
DJ
Reeves
BE
The increased rate of activation of factor XII in late pregnancy can contribute to the increased reactivity of factor VII.
Thromb Haemost
63
1990
349
128
Gordon
EM
Johnson
TR
Ramos
LP
Schmeidler-Sapiro
KT
Enhanced expression of factor XII (Hageman factor) in isolated livers of estrogen-and prolactin-treated rats.
J Lab Clin Med
117
1991
353
129
Farsetti
A
Misiti
S
Citarella
F
Felici
A
Andreoli
M
Fantoni
A
Sacchi
A
Pontecorvi
A
Molecular basis of estrogen regulation of Hageman factor XII gene expression.
Endocrinology
136
1995
5076
130
Schmeidler-Sapiro
KT
Ratnoff
OD
Gordon
EM
Mitogenic effects of coagulation factor XII and factor XIIa on HepG2 cells.
Proc Natl Acad Sci USA
88
1991
4382
131
Gordon
EM
Venkatesan
N
Salazar
R
Tang
H
Schmeidler
K
Buckley
S
Warburton
D
Hall
FL
Factor XII-induced mitogenesis is mediated via a distinct signal transduction pathway that activates a mitogen-activated protein kinase.
Proc Natl Acad Sci USA
93
1996
2174
132
Pixley
RA
Stumpo
LG
Birkmeyer
K
Silver
L
Colman
RW
A monoclonal antibody recognizing an icosapeptide sequence in the heavy chain of human factor XII inhibits surface-catalyzed activation.
J Biol Chem
262
1987
10140
133
Clarke
BJ
Cote
HCF
Cool
DE
Clark-Lewis
I
Saito
H
Pixley
RA
Colman
RW
MacGillivary
RTA
Mapping of a putative surface-binding site of human coagulation factor XII.
J Biol Chem
264
1989
11497
134
Citrella
F
Ravon
DM
Pascucci
B
Felici
A
Fantoni
A
Hack
CE
Structure/function analysis of human factor XII using recombinant deletion mutants. Evidence for an additional region involved in the binding to negatively charged surfaces.
Eur J Biochem
238
1996
240
135
Cochrane
CG
Revak
SD
Wuepper
KD
Activation of Hageman factor in solid and fluid phases. A critical role of kallikrein.
J Exp Med
138
1973
1564
136
Samuel
M
Pixley
RA
Villanueva
MA
Colman
RW
Villanueva
GB
Human factor XII (Hageman factor) autoactivation by dextran sulfate: Circular dichroism, fluorescence, and ultraviolet difference spectroscopic studies.
J Biol Chem
267
1992
19691
137
Pixley
RA
Schapira
M
Colman
RW
Effect of heparin on the inactivation rate of human activated factor XII by antithrombin III.
Blood
66
1985
198
138
Miyata
T
Kawabata
S-I
Iwanaga
S
Takahashi
I
Alving
B
Saito
H
Coagulation factor XII (Hageman factor) Washington DC: Inactive factor XIIa results from Cys571 to Ser substitution.
Proc Natl Acad Sci USA
86
1989
8319
139
Wuillemin
WA
Huber
I
Furlan
M
Lammle
B
Functional characterization of an abnormal factor XII molecule (F XII Bern).
Blood
78
1991
997
140
Cochrane
CG
Revak
SD
Wuepper
KD
Activation of Hageman factor in solid and fluid phases: A critical role of kallikrein.
J Exp Med
138
1973
1564
141
Revak
SD
Cochrane
CG
Griffin
JH
The binding and cleavage characteristics of huma Hageman factor during contact activation: a comparison of normal plasma with plasma deficient in factor XI, prekallikrein or high molecular weight kininogen.
J Clin Invest
59
1977
1167
142
Griffin
JH
The role of surface in the surface-dependent activation of Hageman factor (blood coagulation factor XII).
Proc Natl Acad Sci USA
75
1978
1998
143
Kirby
E
McDevitt
PJ
The binding of bovine factor XII to kaolin.
Blood
61
1983
652
144
Wiggins
RC
Cochrane
CG
The autoactivation of rabbit Hageman factor.
J Exp Med
150
1979
1122
145
Miller
G
Silverberg
M
Kaplan
AP
Autoactivability of human Hageman factor.
Biochem Biophys Res Commun
92
1980
803
146
Silverberg
M
Dunn
JT
Garen
L
Kaplan
AP
Autoactivation of human Hageman factor: Demonstration utilizing a synthetic substrate.
J Biol Chem
255
1980
7281
147
Dunn
JT
Silverberg
M
Kaplan
AP
The cleavage and formation of activated human Hageman factor by autodigestion and by kallikrein.
J Biol Chem
257
1982
1779
148
Espana
F
Ratnoff
OD
Activation of Hageman factor (factor XII) by sulfatides and other agents in the absence of plasma proteases.
J Lab Clin Med
102
1983
31
149
Tankersley
DL
Finlayson
JS
Kinetics of activation and autoactivation of human factor XII.
Biochemistry
23
1984
273
150
Schousboe
I
Contact activation in human plasma is triggered by zinc ion modulation of factor XII (Hageman factor).
Blood Coagul Fibrinolysis
4
1993
671
151
Bernardo
MM
Day
DE
Olson
ST
Shore
JD
Surface-independent acceleration of factor XII activation by zinc ions. I. Kinetic characterization of the metal ion rate enhancement.
J Biol Chem
268
1993
12468
152
Bernardo
MM
Day
DE
Halvorson
HR
Olson
ST
Shore
JD
Surface-independent acceleration of factor XII activation by zinc ions. II. Direct binding and fluorescence studies.
J Biol Chem
268
1993
12477
153
Rojkjaer
R
Schousboe
I
Identification of the Zn2+ binding sites in factor XII and its activation derivatives.
Eur J Biochem
247
1997
491
154
Wiggins
RC
Loskutoff
DJ
Cochrane
CG
Griffin
JH
Activation of rabbit Hageman factor by homogenates of cultured rabbit endothelial cells.
J Clin Invest
65
1980
197
155
Bagdasarian
A
Talamo
RC
Colman
RW
Isolation of the high molecular weight activators of prekallikrein.
J Biol Chem
248
1973
7742
156
Kaplan
AP
Austen
FK
A prealbumin activator of prekallikrein.
J Immunol
105
1970
802
157
Wuepper
KD
Tucker
ES III
Cochrane
CG
Plasma Kinin system: Proenzyme components.
J Immunol
105
1970
1307
158
Cochrane
CG
Griffin
JH
Molecular assembly in the contact phase of the Hageman factor system.
Am J Med
67
1979
657
159
Kaplan
AP
Austen
FK
A prealbumin activator of prekallikrein. II. Derivation of activators of prekallikrein from active Hageman factor by digestion with plasmin.
J Exp Med
133
1971
696
160
Revak
SD
Cochrane
CG
Bouma
BN
Griffin
JH
Surface and fluid phase activities of two forms of activated Hageman factor produced during contact activation of plasma.
J Exp Med
147
1978
719
161
Revak
SD
Cochrane
CG
The relationship of structure and function in human Hageman factor. The association of enzymatic binding activities with separate regions of the molecule.
J Clin Invest
57
1976
852
162
Forbes
CO
Pensky
J
Ratnoff
OD
Inhibition of activated Hageman factor and activated plasma thromboplastin antecedent by purified C1-inactivator.
J Lab Clin Med
76
1970
809
163
Schreiber
AD
Kaplan
AD
Austen
FK
Inhibition by C1-INH of Hageman factor fragment activation of coagulation, fibrinolysis and kinin-generation.
J Clin Invest
52
1973
1402
164
de Agostini
A
Lijnen
HR
Pixley
RA
Colman
RW
Schapira
M
Inactivation of factor XII active fragment in normal plasma: Predominant role of C1-inhibitor.
J Clin Invest
73
1984
1542
165
Pixley
RA
Schapira
M
Colman
RW
The regulation of human factor XIIa by plasma proteinase inhibitors.
J Biol Chem
260
1985
1723
166
Pixley
RA
Schmaier
AH
Colman
RW
Effect of negatively charged activating compounds on inactivation of factor XIIa by C1 inhibitor.
Arch Biochem Biophys
256
1987
490
167
Stead
NW
Kaplan
AP
Rosenberg
RD
The inhibition of human activated Hageman factor (HF ) by human antithrombin-heparin cofactor (AT).
J Biol Chem
251
1976
6481
168
Pixley
RA
Colman
RW
Effect of heparin on the inactivation rate of human activated factor XII by antithrombin III.
Blood
66
1985
198
169
Berrettini
M
Schleef
RR
Espana
F
Loskutoff
DJ
Griffin
JH
Interaction of type 1 plasminogen activator inhibitor with the enzymes of the contact activation system.
J Biol Chem
264
1989
11738
170
Kleniewski
J
Donaldson
VH
Endothelial cells produce a substance that inhibits contact activation of coagulation by blocking the activation of Hageman factor.
Proc Natl Acad Sci USA
90
1993
198
171
Greengard
JS
Griffin
JH
Receptors for high molecular weight kininogen on stimulated washed human platelets.
Biochemistry
23
1984
6863
172
van Iwaarden
F
de Groot
PG
Sixma
JJ
Berrettini
M
Bouma
BN
High-molecular weight kininogen is present in cultured human endothelial cells: Localization, isolation, and characterization.
Blood
71
1988
1268
173
Kerbiriou-Nabias
DM
Garcia
FO
Larrieu
MJ
Radioimmunoassays of human high and low molecular weight kininogens in plasmas and platelets.
Br J Haematol
56
1984
273
174
Figueroa
CD
Henderson
LM
Kaufmann
J
DeLa
Cadena RA
Colman
RW
Muller-Esterl
W
Bhoola
KD
Immunovisualization of high (HK) and low (LK) molecular weight kininogens on isolated human neutrophils.
Blood
79
1992
759
175
Gustafson
EJ
Lukasiewicz
H
Wachtfogel
YT
Norton
KJ
Schmaier
AH
Niewiarowski
S
Colman
RW
High molecular weight kininogen inhibits fibrinogen binding to cytoadhesins of neutrophils and platelets.
J Cell Biol
109
1989
377
176
Berrettini
M
Schleef
RR
Heeb
MJ
Hopmeier
P
Griffin
JH
Assembly and expression of an intrinsic factor IX activator complex on the surface of cultured human endothelial cells.
J Biol Chem
267
1992
19833
177
Jencks
WP
On the attribution and additivity of binding energies.
Proc Natl Acad Sci USA
78
1981
4046
178
Villanueva
GB
Leung
L
Bradford
H
Colman
RW
Conformation of high molecular weight kininogen: Effects of kallikrein and factor XIa cleavage.
Biochem Biophys Res Commun
158
1989
72
179
Edgell
CJS
McDonald
CC
Graham
JB
Permanent cell lines expressing human factor VIII-related antigen established by hybridization.
Proc Natl Acad Sci USA
80
1983
3734
180
Herwald
H
Dedio
J
Kellner
R
Loos
M
Mueller-Esterl
W
Isolation and characterization of the kininogen-binding protein p33 from endothelial cells: Identity with the gC1q receptor.
J Biol Chem
271
1996
13040
181
Ghebrehiwet
B
Lim
BL
Peerschke
EI
Willis
AC
Reid
KB
Isolation, cDNA cloning, and overexpression of a 33-kD cell surface glycoprotein that binds to the globular “heads” of C1q.
J Exp Med
179
1994
1809
182
Joseph
K
Ghebrehiwet
B
Peerschke
EIB
Reid
KBM
Kaplan
AP
Identification of the zinc-dependent endothelial cell binding protein for high molecular weight kininogen and factor XII: Identity with the receptor that binds to the globular “heads” of C1q (gC1qR).
Proc Natl Acad Sci USA
93
1996
8552
183
Dedio J, Mueller-Esterl W. Kininogen binding protein p33/gC1qR is localized in the vesicular fraction of endothelial cells. FEBS Lett 399:255, 1996
184
Reddigari
SR
Shibayama
Y
Brunnee
T
Kaplan
AP
Human Hageman factor (factor XII) and high molecular weight kininogen compete for the same binding site on human umbilical vein endothelial cells.
J Biol Chem
268
1993
11982
185
Colman RW, Pixley RA, Najamunnisa S, Yan W-Y, Wang J, Mazar A, McCrae KR: High molecular weight kininogen binds to the vitronectin binding domain(s) on the endothelial cell receptor. Circulation 94:I-42, 1996 (abstr)
186
Wei
Y
Lukashev
M
Simon
DI
Bodary
SC
Rosenberg
S
Doyle
MV
Chapman
HA
Regulation of integrin function by the urokinase receptor.
Science
273
1996
1551
186a
Sheng N, Fairbanks M, Henrikson R, Canziani G, Chaiken I, Moser D, Colman RW: The integrin, CD11b/CD18 (Mac-1) is a major cell receptor for high molecular weight kininogen (HK). FASEB J 11:19920, 1997 (abstr)
187
Hasan AAK, Zisman T, Schmaier AH: Cytokeratin 1 is the major cell receptor for kininogens. Thromb Haemost 77:141, 1997 (abstr, suppl 1)
188
Hembrough
TA
Vasudevan
J
Allietta
MM
Glass
WF II
Gonias
SL
A cytokeratin 8-like protein with plasminogen-binding activity is present on the external surface of hepatocytes, HepG2 cells, and breast carcinoma cell line.
J Cell Sci
108
1995
1071
189
Hembrough
TA
Li
L
Gonias
SL
Cell-surface cytokeratin 8 is the major plasminogen receptor on breast cancer cells and is required for the accelerated activation of cell-associated plasminogen by tissue-type plasminogen activator.
J Biol Chem
271
1996
25684
190
Nishikawa
K
Shibayama
Y
Kuna
P
Calcaterra
E
Kaplan
AP
Reddigari
SR
Generation of vasoactive peptide bradykinin from human umbilical vein endothelium-bound high molecular weight kininogen by plasma kallikrein.
Blood
80
1992
1980
191
Sinha
D
Seaman
FS
Koshy
A
Knight
LC
Walsh
PN
Blood coagulation factor XIa binds specifically to a site on activated human platelets distinct from that for factor XI.
J Clin Invest
73
1984
1550
192
Walsh
PN
Sinha
D
Koshy
A
Seaman
FS
Bradford
H
Functional characterization of platelet-bound factor XIa: Retention of factor XIa activity on the platelet surface.
Blood
68
1986
225
193
Loza
JP
Gurewich
V
Johnstone
M
Pannell
R
Platelet-bound prekallikrein promotes pro-urokinase-induced clot lysis: A mechanism for targeting the factor XII dependent intrinsic pathway of fibrinolysis.
Thromb Haemost
71
1994
347
194
Kaplan
AP
Kay
AR
Austen
KF
A prealbumin activator of prekallikrein II. Appearance of chemotactic activity for human neutrophils by the conversion of prekallikrein to kallikrein.
J Exp Med
135
1972
81
195
Schapira
M
Despland
E
Scott
CF
Boxer
LA
Colman
RW
Purified human plasma kallikrein aggregates human blood neutrophils.
J Clin Invest
69
1982
1199
196
Wachtfogel
YT
Kucich
U
James
HL
Scott
CF
Schapira
M
Zimmerman
M
Cohen
AB
Colman
RW
Human plasma kallikrein releases neutrophil elastase during blood coagulation.
J Clin Invest
72
1983
1672
197
Zimmerli
W
Huber
I
Bouma
BN
Lammle
B
Purified human plasma kallikrein does not stimulate but primes neutrophils for superoxide production.
Thromb Haemost
62
1989
1121
198
Plow
EF
Leukocyte elastase release during blood coagulation: A potential mechanism for activation of the alternative fibrinolytic pathway.
J Clin Invest
69
1982
564
199
Rebuck
JW
The skin window as a monitor of leukocytic functions in contact activation factor deficiencies in man.
Am J Clin Pathol
79
1983
405
200
Colman
RW
Wachtfogel
YT
Kucich
U
Weinbaum
G
Hahn
S
Pixley
RA
Scott
CF
DeAgostini
A
Burger
D
Schapira
M
Effect of cleavage of the heavy chain of human plasma kallikrein on its functional properties.
Blood
65
1985
311
201
Wachtfogel
YT
Pixley
RA
Kucich
U
Abrams
W
Weinbaum
G
Schapira
M
Colman
RW
Purified plasma factor XIIa aggregates human neutrophils and causes degranulation.
Blood
67
1986
1731
202
Chien
P
Pixley
RA
Stumpo
LG
Colman
RW
Schreiber
AD
Modulation of the human monocyte binding site for monomeric immunoglobulin G by activated Hageman factor.
J Clin Invest
82
1988
1554
203
Chien P, Pixley RA, Ruiz P, Colman RW, Schreiber AD: Modulation of the human monocyte FcgR1 by activated Hageman factor: Mapping the functional XIIa site. Blood 76:178a, 1990 (abstr, suppl 1)
204
Toossi
Z
Sedor
JR
Mettler
MA
Everson
B
Young
T
Ratnoff
OD
Induction of expression of monocyte interleukin 1 by Hageman factor (factor XII).
Proc Natl Acad Sci USA
89
1992
11969
205
Crutchley
DJ
Ryan
JW
Ryan
US
Fisher
GH
Bradykinin-induced release of prostacyclin and thromboxanes from bovine pulmonary artery endothelial cells. Studies with lower homologs and calcium antagonists.
Biochim Biophys Acta
751
1983
99
206
Hong
SL
Effect of bradykinin and thrombin on prostacyclin synthesis in endothelial cells from calf and pig aorta and human umbilical cord vein.
Thromb Res
18
1980
787
207
Holland
JA
Pritchard
KA
Pappolla
MA
Wolin
MS
Rogers
NJ
Stemerman
MB
Bradykinin induces superoxide anion release from human endothelial cells.
J Cell Physiol
143
1990
21
208
Smith
D
Gilbert
M
Owen
WG
Tissue plasminogen activator release in vivo in response to vasoactive agents.
Blood
66
1983
835
209
Brown
NJ
Nadeau
JH
Vaughan
DE
Selective stimulation of tissue-type plasminogen activator (t-PA) in vivo by infusion of bradykinin.
Thromb Haemost
77
1997
522
210
Palmer
RMJ
Ferrige
AG
Moncada
S
Nitric oxide release accounts for the biologic activity of endothelium-derived relaxing factor.
Nature
327
1987
524
211
Nakashima
M
Mombouli
JV
Taylor
AA
Vanhoutte
PM
Endothelium-dependent hyperpolarization caused by bradykinin in human coronary arteries.
J Clin Invest
92
1993
2867
212
Boulanger
C
Schini
VB
Moncada
S
Vanhoutte
PM
Stimulation of cyclic GMP production in cultured endothelial cells of the pig by bradykinin, adenosine diphosphate, calcium ionophore A23187 and nitric oxide.
Br J Pharmacol
101
1990
152
213
Schini
VB
Boulanger
C
Regoli
D
Vanhoutte
PM
Bradykinin stimulates the production of cyclic GMP via activation of B2 kinin receptors in cultured porcine aortic endothelial cells.
J Pharmacol Exp Ther
252
1990
581
214
Busse
R
Mulsch
A
Induction of nitric oxide synthase by cytokines in vascular smooth muscle cells.
FEBS Lett
275
1990
87
215
Imai
T
Hirata
Y
Kanno
K
Marumo
F
Induction of nitric oxide synthase by cyclic AMP in rat vascular smooth muscle cells.
J Clin Invest
93
1994
543
216
Dixon BS, Breckon R, Fortune J, Vavrek RJ, Stewart JM, Marzec-Calvert R, Linas SL: Effects of kinins on cultured arterial smooth muscle. Am J Physiol 258:C299, 1990
217
Dixon BS, Sharma RV, Dickerson T, Fortune J: Bradykinin and angiotensin II: Activation of protein kinase C in arterial smooth muscle. Am J Physiol 266:C1406, 1994
218
McEachern
AE
Shelton
ER
Bhakta
S
Obernolte
R
Bach
C
Zuppan
P
Fujisaki
J
Aldrich
RW
Jarnagin
K
Expression cloning of a rat B2 bradykinin receptor.
Proc Natl Acad Sci USA
88
1991
7724
219
Menke
JG
Borkowski
JA
Bierilo
K
MacNeil
T
Derrick
A
Schneck
KA
Ransom
RW
Strader
CD
Linemayer
DL
Hess
JF
Expression cloning of a human B1 bradykinin receptor.
J Biol Chem
269
1994
21583
220
Madeddu
P
Parpaglia
PP
Demontis
MP
Varoni
MV
Fattaccio
MC
Anania
V
Glorioso
N
Early blockade of bradykinin B2-receptors alters the adult cardiovascular phenotype in rats.
Hypertension
25
1995
453
221
Wiemer
G
Scholkens
BA
Linz
W
Endothelial protection by converting enzyme inhibitors.
Cardiovasc Res
28
1994
166
222
Parratt
JR
Cardioprotection by angiotensin converting enzyme inhibitors-the experimental evidence.
Circ Res
28
1994
183
223
Linz
W
Wiemer
G
Scholkens
BA
ACE-inhibition induces NO-formation in cultured bovine endothelial cells and protects isolated ischemic rat hearts.
Mol Cell Cardiol
24
1992
909
224
Linz W, Scholkens BA: Role of bradykinin in the cardiac effects of angiotensin-converting enzyme inhibitors. J Cardiovasc Pharm 20:S83, 1992 (suppl 9)
225
Gohlke
P
Linz
W
Scholkens
BA
Kuwer
I
Bartenbach
S
Schnell
A
Unger
T
Angiotensin-converting enzyme inhibition improves cardiac function. Role of bradykinin.
Hypertension
23
1994
411
226
O'Sullivan
JB
Harrap
SB
Resetting blood pressure in spontaneously hypertensive rats. The role of bradykinin.
Hypertension
25
1995
162
227
Siebeck
M
Cheronis
JC
Fink
E
Kohl
J
Spies
B
Spannagl
M
Jochum
M
Fritz
H
Dextran sulfate activates contact system and mediates arterial hypotension via B2 kinin receptors.
J Appl Physiol
77
1994
2675
228
Wang
J
Xiong
W
Yang
Z
Davis
T
Dewey
MJ
Chao
J
Chao
L
Human tissue kallikrein induces hypotension in transgenic mice.
Hypertension
23
1994
236
229
Ma
JX
Yang
Z
Chao
J
Chao
L
Intramuscular delivery of rat kallikrein-binding protein gene reverses hypotension in transgenic mice expressing human tissue kallikrein.
J Biol Chem
270
1995
451
230
Chao
J
Tillman
DM
Wang
MY
Margolius
HS
Chao
L
Identification of a new tissue-kallikrein-binding protein.
Biochem J
239
1986
325
231
Chao
J
Chai
KX
Chen
LM
Xiong
W
Chao
S
Woodley-Miller
C
Wang
LX
Chao
L
Tissue kallikrein-binding protein is a serpin. I. Purification, characterization, and distribution in normotensive and spontaneously hypertensive rats.
J Biol Chem
265
1990
16394
232
Zhou
GX
Chao
L
Chao
J
Kallistatin: A novel human tissue kallikrein inhibitor. Purification, characterization, and reactive center sequence.
J Biol Chem
267
1992
25873
233
Wang
C
Chao
L
Chao
J
Direct gene delivery of human tissue kallikrein reduces blood pressure in spontaneously hypertensive rats [see comments].
J Clin Invest
95
1995
1710
234
Xiong
W
Chao
J
Chao
L
Muscle delivery of human kallikrein gene reduces blood pressure in hypertensive rats.
Hypertension
25
1995
715
235
Puri
RN
Gustafson
EJ
Zhou
FX
Bradford
H
Colman
RF
Colman
RW
Inhibition of thrombin-induced platelet aggregation by high molecular weight kininogen.
Trans Assoc Am Phys
100
1987
232
236
Coller
BS
Effects of tertiary amine local anesthetics on von Willebrand factor-dependent platelet function: Alteration of membrane reactivity and degradation of GPIb by a calcium-dependent protease(s).
Blood
60
1982
731
237
Puri
RN
Matsueda
R
Umeyama
H
Bradford
HN
Colman
RW
Modulation of thrombin-induced platelet aggregation by inhibition of calpain by a synthetic peptide derived from the thiol-protease inhibitory sequence of kininogens and S-(3-nitro-2-pyridinesulfenyl)-cysteine.
Biochem Biophys Res Commun
162
1989
1017
238
Matsueda
R
Umeyama
H
Puri
RN
Bradford
HN
Colman
RW
Design and synthesis of a kininogen-based selective inhibitor of thrombin-induced platelet aggregation.
Pept Res
7
1994
32
239
Charo
IF
Feinman
RD
Detwiler
TC
Interrelations of platelet aggregation and secretion.
J Clin Invest
60
1977
866
240
Schmaier
AH
Meloni
FJ
Nawarawong
W
Jiang
YP
PPACK-thrombin is a noncompetitive inhibitor of alpha-thrombin binding to human platelets.
Thromb Res
67
1992
479
241
Bradford
HN
DeLa
Cadena RA
Kunapuli
SP
Dong
JF
Lopez
JA
Colman
RW
Human kininogens regulate thrombin binding to platelets through the GPIb-IX complex.
Blood
90
1997
1508
242
Majima
M
Sunahara
N
Harada
Y
Katori
M
Detection of the degradation products of bradykinin by enzyme immunoassays as markers for the release of kinin in vivo.
Biochem Pharmacol
45
1993
559
243
Shima
C
Majima
M
Katori
M
A stable metabolite, Arg-Pro-Pro-Gly-Phe, of bradykinin in the degradation pathway in human plasma.
Jpn J Pharmacol
60
1992
111
244
Niewiarowski
S
Prou-Wartelle
O
Role of the contact factor (Hageman factor) in fibrinolysis.
Thromb Diath Haemorrh
3
1959
593
245
Colman
RW
Activation of plasminogen by human plasma kallikrein.
Biochem Biophys Res Commun
35
1969
273
246
Mandle
RJ Jr
Kaplan
AP
Human plasma prekallikrein: Mechanism of activation by Hageman factor and participation in Hageman-factor-dependent fibrinolysis.
J Biol Chem
252
1977
6097
247
Goldsmith
G
Saito
H
Ratnoff
OD
The activation of plasminogen by Hageman FXII (factor XII) and Hageman factor fragments.
J Clin Invest
62
1978
54
248
Mandle
RJ Jr
Kaplan
AP
Hageman-factor-dependent fibrinolysis: Generation of fibrinolytic activity by the interaction of human activated factor XI and plasminogen.
Blood
54
1979
850
249
Gurewich
V
Johnstone
M
Loza
JP
Pannell
R
Pro-urokinase and prekallikrein are both associated with platelets. Implications for the intrinsic pathway of fibrinolysis and for therapeutic thrombolysis.
FEBS Lett
318
1993
317
250
Lin
Y
Harris
RB
Yan
W
McCrae
KR
Zhang
H Colman RW
High molecular weight kininogen peptides inhibit the formation of kallikrein on endothelial cell surfaces and subsequent urokinase-dependent plasmin formation.
Blood
90
1997
690
251
Higazi
A
Cohen
RL
Henkin
J
Kniss
D
Schwartz
BS
Cines
DB
Enhancement of the enzymatic activity of single-chain urokinase plasminogen activator by soluble urokinase receptor.
J Biol Chem
21
1995
17375
252
Vroman
L
Adams
A
Possible involvement of fibrinogen and proteolysis in surface activation: A study with the recording ellipsometer.
Thromb Diath Haemorrh
18
1967
510
253
Schmaier
AH
Silver
L
Adams
AL
Fischer
GC
Munoz
PC
Vroman
L
Colman
RW
The effect of high molecular weight kininogen on surface-adsorbed fibrinogen.
Thromb Res
33
1984
51
254
Brash
JL
Scott
CF
ten Hove
P
Wojciechowski
P
Colman
RW
Mechanism of transient adsorption of fibrinogen from plasma to solid surfaces: Role of the contact and fibrinolytic systems.
Blood
71
1988
932
255
Yung
LL
Lim
F
Khan
MMH
Kunapuli
SP
Rick
L
Colman
RW
Cooper
SL
Neutrophil adhesion on surfaces preadsorbed with high molecular weight kininogen under well-defined flow conditions.
Immunopharmacology
32
1996
9
256
Donaldson
VH
Evans
RR
Biochemical abnormality in hereditary angioneurotic edema.
Am J Med
35
1963
37
257
Ratnoff
OD
Lepow
IH
Some properties of an esterase derived from preparations of the first component of complement.
J Exp Med
106
1955
327
258
Landerman NS Hereditary angioneurotic edema. I. Case reports and review of the literature. J Allergy Clin Immunol 33:316, 1962
259
Schapira
M
Sliver
LD
Scott
CF
Schmaier
AH
Prograis
L
Curd
J
Colman
RW
Prekallikrein activation and high molecular weight kininogen consumption in hereditary angioedema.
N Engl J Med
308
1983
1050
260
Cugno
M
Hack
CE
de Boer
JP
Eerenberg
AJ
Agostini
A
Cicardi
M
Generation of plasmin during acute attacks of hereditary angioedema.
J Lab Clin Med
121
1993
38
261
Berrettini
M
Lammle
B
White
T
Heeb
MJ
Schwarz
HP
Zuraw
B
Curd
J
Griffin
JH
Detection of in vitro and in vivo cleavage of high molecular weight kininogen in human plasma by immunblotting with monoclonal antibodies.
Blood
68
1986
455
262
Fields
AP
Ghebrehiwet
B
Kaplan
AP
Kinin formation in hereditary angioedema plasma: Evidence aginst kinin derivation from C2 and in support of “spontaneous” formation of bradykinin.
J Allergy Clin Immunol
72
1983
54
263
Rapaport
S
Aas
K
Owen
PA
Effect of glass upon the activation of various plasma clotting factors.
J Clin Invest
34
1955
9
264
Gjonnaess
H
Cold-promoted activation of factor VII. III. Relation to the kallikrein system.
Thromb Diath Haemorrh
28
1972
182
265
Seligsohn
U
Osterud
B
Brown
SF
Griffin
JH
Rapaport
S
Activation of human factor VII in plasma and in purified systems.
J Clin Invest
64
1979
239
266
Weiss
R
Kaplan
AP
The effect of C1 inhibitor upon Hageman factor autoactivation.
Blood
68
1986
239
267
Kalter
ES
Daha
MR
Verhoef
J
Bouma
BN
Activation and inhibition of Hageman factor-dependent pathways and the complement system in uncomplicated bacteremia or bacterial shock.
J Infect Dis
151
1985
1019
268
Colman
RW
The role of plasma proteases in septic shock.
N Engl J Med
320
1989
1207
269
Carvalho
AC
DeMarinis
S
Scott
CF
Silver
LD
Schmaier
AH
Colman
RW
Activation of the contact system of plasma proteolysis in the adult respiratory distress syndrome.
J Lab Clin Med
112
1988
270
270
Mason
JW
Colman
RW
The role of Hageman factor in disseminated intravascular coagulation induced by septicemia, neoplasia, or liver disease.
Thromb Diath Haemorrh
26
1971
325
271
O'Donnell
TF
Clowes
GH
Talamo
RC
Colman
RW
Kinin activation in the blood of patients with sepsis.
Surg Gynecol Obstet
143
1976
539
272
Colman
RW
Edelman
R
Scott
CF
Gilman
RH
Plasma kallikrein activation and inhibition during typhoid fever.
J Clin Invest
61
1978
287
273
Schapira
M
Gardaz
JP
Py
P
Lew
PD
Perrin
LH
Suter
PM
Prekallikrein activation in the adult respiratory distress syndrome.
Bull Eur Physiopathol Respir
21
1985
237
274
Rao
AK
Schapira
M
Clements
ML
Niewiarowski
S
Budzynski
AZ
Schmaier
AH
Harpel
PC
Blackwelder
WC
Scherrer
JR
Sobel
E
A prospective study of platelets and plasma proteolytic systems during the early stages of Rocky Mountain spotted fever.
N Engl J Med
318
1988
1021
275
Nuijens
JH
Huijbregts
CC
Eerenberg
Belmer AJ
Abbink
JJ
Strack van Schijndel
RJ
Felt
Bersma RJ
Thijs
LG
Hack
CE
Quantification of plasma factor XIIa-Cl(−)-inhibitor and kallikrein-Cl(−)-inhibitor complexes in sepsis.
Blood
72
1988
1841
276
Kaufman
N
Page
JD
Pixley
RA
Schein
R
Schmaier
AH
Colman
RW
Alpha 2-macroglobulin-kallikrein complexes detect contact system activation in hereditary angioedema and human sepsis.
Blood
77
1991
2660
277
DeLa
Cadena RA
Suffredini
AF
Page
JD
Pixley
RA
Kaufman
N
Parrillo
JE
Colman
RW
Activation of the kallikrein-kinin system after endotoxin administration to normal human volunteers.
Blood
81
1993
3313
278
Pixley
RA
DeLa
Cadena RA
Page
JD
Kaufman
N
Wyshock
EG
Chang
A
Taylor
FB Jr
Colman
RW
Activation of the contact system in lethal hypotensive bacteremia in a baboon model.
Am J Pathol
140
1992
1
279
Pixley
RA
DeLa
Cadena RA
Page
JD
Kaufman
N
Wyshock
EG
Chang
A
Taylor
FB Jr
Colman
RW
The contact system contributes to hypotension but not disseminated intravascular coagulation in lethal bacteremia: In vivo use of a monoclonal anti-factor XII antibody to block contact activation in baboons.
J Clin Invest
91
1993
61
280
Colman
RW
Scott
CF
Pixley
RA
DeLa
Cadena RA
Effect of heparin on the inhibition of the contact system enzymes.
Ann NY Acad Sci
556
1989
95
281
Wachtfogel
YT
Harpel
PC
Edmunds
LH Jr
Colman
RW
Formation of C1s-C1-inhibitor, kallikrein-C1-inhibitor, and plasmin-alpha 2-plasmin-inhibitor complexes during cardiopulmonary bypass.
Blood
73
1989
468
282
Wachtfogel
YT
Kucich
U
Hack
CE
Gluszko
P
Niewiarowski
S
Colman
RW
Edmunds
LH Jr
Aprotinin inhibits the contact, neutrophil and platelet activation systems during simulated extracorporeal perfusion.
J Thorac Cardiovasc Surg
106
1993
1
283
Wachtfogel YT, Hack CE, Nuijens JH, Kettner C, Reilly TM, Knabb RM, Bischoff R, Tschesche H, Wenzel H, Kucich U, Edmunds LH Jr, Colman RW: Selective kallikrein inhibitors alter human neutrophil elastase release during extracorporeal circulation. Am J Physiol 268:H1352, 1995
284
DeLa Cadena RA, Laskin KJ, Pixley RA, Sartor RB, Schwab JH, Back N, Bedi GS, Fisher RS, Colman RW: Role of kallikrein-kinin system in pathogenesis of bacterial cell wall-induced inflammation. Am J Physiol 260:G213, 1991
285
DeLa
Cadena RA
Stadnicki
A
Uknis
AB
Sartor
RB
Kettner
CA
Adam
A
Colman
RW
Inhibition of plasma kallikrein prevents peptidoglycan-induced arthritis in the Lewis rat.
FASEB J
9
1995
446
286
Sartor
RB
DeLa
Cadena RA
Green
KD
Stadnicki
A
Davis
SW
Schwab
JH
Adam
AA
Raymond
P
Colman
RW
Selective kallikrein-kinin system activation in inbred rats differentially susceptible to granulomatous enterocolitis.
Gastroenterology
110
1996
1467
287
Stadnicki
A
DeLa
Cadena RA
Sartor
RB
Bender
D
Kettner
CA
Rath
HC
Adam
A
Colman
RW
Selective plasma kallikrein inhibitor attenuates acute intestinal inflammation in Lewis rat.
Dig Dis Sci
41
1996
912
288
Jansen
PM
Pixley
RA
Brouwer
M
DeJong
IW
Chang
AK
Hack
CE
Taylor
FB Jr
Colman
RW
Inhibition of factor XII in septic baboons attenuates the activation of complement and fibrinolytic systems and reduces the release of interleukin-6 and neutrophil elastase.
Blood
87
1996
2337
289
Goodnough
LT
Saito
H
Ratnoff
OD
Thrombosis or myocardial infarction in congenital clotting factor abnormalities and chronic thrombocytopenias: A report of 21 patients and a review of 50 previously reported cases [Review].
Medicine (Baltimore)
62
1983
248
290
Mannhalter
C
Fisher
M
Hopmeier
P
Deutch
E
Factor XII activity and antigen concentrations in patients suffering from recurrent thrombosis.
Fibrinolysis
1
1987
259
291
Lammle
B
Wuillemin
WA
Huber
I
Krauskopf
M
Zurcher
C
Pflugshaupt
R
Furlan
M
Thromboembolism and bleeding tendency in congenital factor XII deficiency: A study on 74 subjects from 14 Swiss families.
Thromb Haemost
65
1991
117
292
Halbmayer
WM
Mannhalter
C
Feichtinger
C
Rubi
K
Fischer
M
The prevalence of factor XII deficiency in 103 orally anticoagulated outpatients suffering from recurrent venous and/or arterial thromboembolism.
Thromb Haemost
68
1992
285
293
von Kanel
R
Wuillemin
WA
Furlan
M
Lammle
B
Factor XII clotting activity and antigen levels in patients with thromboembolic disease.
Blood Coagul Fibrinolysis
3
1992
555
294
Jespersen
J
Munkvad
S
Pedersen
OD
Gram
J
Kluft
C
Evidence for a role of factor XII-dependent fibrinolysis in cardiovascular diseases.
Ann NY Acad Sci
667
1992
454
295
Munkvad
S
Jespersen
J
Gram
J
Kluft
C
Long-lasting depression of the factor XII-dependent fibrinolytic system in patients with myocardial infarction undergoing thrombolytic therapy with recombinant tissue-type plasminogen activator: A randomized placebo-controlled study.
J Am Coll Cardiol
17
1991
957
Sign in via your Institution