SlideShare una empresa de Scribd logo
1 de 7
Descargar para leer sin conexión
Journal of Thrombosis and Haemostasis, 5: 2323–2329



    REVIEW ARTICLE


The plasma kallikrein–kinin system: its evolution from contact
activation
A . H . S C H M A I E R and K . R . M C C R A E
Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University and University Hospitals Case Medical Center,
Cleveland, OH, USA



To cite this article: Schmaier AH, McCrae KR. The plasma kallikrein–kinin system: its evolution from contact activation. J Thromb Haemost 2007; 5:
2323–9.



                                                                           review will outline physiologic activities of the plasma KKS
Summary. The plasma kallikrein–kinin system consists of the                that are not emphasized in other recent reviews [1,2].
proteins factor XII (FXII), prekallikrein (PK), and high                      FXII deficiency (Hageman trait) was discovered by Ratnoff
molecular weight kininogen. It was first recognized as a sur-               and Colopy in an individual who had prolonged blood clotting
face-activated coagulation system that is activated when blood             times without bleeding [3]. Activation of FXII results in FXI
or plasma interacts with artificial surfaces. Although surface-             activation, giving rise to the coagulation cascade [4]. As result
activated contact activation occurs in vivo in the case of tissue          of elucidating non-FXII-deficient etiologies for isolated pro-
destruction or a developing thrombus, the physiologic basis for            longed activated partial thromboplastin times (APTT), plasma
the activation and function of this system has not been                    PK and HK were discovered [5,6]. These proteins influence the
delineated. New investigations indicate that there is a proteo-             200 million surface-activated coagulation tests, APTTs and
lytic pathway on cells for PK activation independent of FXII.              activated clotting times performed annually in the USA. New
This pathway for PK with subsequent FXII activation indicates              interest in FXII has arisen since it has been observed that FXII-
physiologic activities. These activities include blood pressure            deficient mice have reduced thrombus compared to the wild
regulation and modulation of thrombosis risk independently of              type [7]. Furthermore, bradykinin (BK) B2 receptor (B2R)-
hemostasis. Furthermore, they include regulation of endothelial            deficient mice also have reduced thrombosis risk [8]. C1 esterase
cell proliferation, angiogenesis and apoptosis through a cellular-         inhibitor (C1INH), the SERPIN inhibitor of the enzymes of
based, outside-in signaling system. The present characteriza-              this system, accounts for 90% of inhibition of FXIIa and 50%
tions of this system, which incorrectly had been thought to                of inhibition of plasma kallikrein [9,10] (Table 2). Although
initiate coagulation, represent an evolution of understanding in           plasminogen activator inhibitor-1 and protein C inhibitor,
this field.                                                                 mole for mole, may be more potent inhibitors of plasma
                                                                           kallikrein than C1INH, the intravascular concentration of
Introduction                                                               C1INH is highest, making it the most important (Table 2).
                                                                           C1INH deficiency is the etiology of hereditary angioedema, a
Appreciation of the plasma kallikrein–kinin system (KKS) has
                                                                           disorder associated with unregulated BK formation, producing
grown. The KKS consists of two zymogens, factor XII (FXII)
                                                                           secondary angioedema in humans and mice [11].
and prekallikrein (PK), and one substrate/cofactor, high
molecular weight kininogen (HK) (Table 1). These proteins
influence surface-activated in vitro coagulation assays, but                Contact activation of FXII and the proteins of the plasma
deficiencies are not associated with bleeding. Recent studies               KKS
indicate activities in vascular biology, including modulation of
                                                                           The three proteins (FXII, PK, and HK) of the plasma KKS are
thrombosis risk independently of blood coagulation. This
                                                                           called Ôcontact factorsÕ because, until recently (see next section),
                                                                           there was no known mechanism for their initiation of activation
                                                                           other than FXII autoactivation on surfaces [2,3]. Autoactiva-
Correspondence: Alvin H. Schmaier, Case Western Reserve
                                                                           tion is the event where zymogen FXII becomes an enzyme in the
University, University Hospital Case Medical Center, Division of
                                                                           presence of a negatively charged surface, a process twentynine-
Hematology and Oncology, 10900 Euclid Avenue WRB2-130,
Cleveland, OH 44106-7284, USA.
                                                                           fold less efficient than activation by plasma kallikrein [12]
Tel.: +1 216 368 1172; fax: +1 216 368 3014; e-mail:                       (Table 1). The biochemistry of this phenomenon is not under-
schmaier@case.edu                                                          stood, but recent studies using sum frequency generation
                                                                           vibrational spectroscopy indicate that FXII autoactivation at
Received 3 August 2007, accepted 14 September 2007                         the molecular level occurs by imposing specific orientation and

Ó 2007 International Society on Thrombosis and Haemostasis
2324 A. H. Schmaier and K. R. McCrae

Table 1 Enzymes/substrates of the plasma kallikrein–kinin system
Enzyme                                      Substrate                               Kinetics                                             Reference

a-Factor (F) XIIa                           Prekallikrein                           1.8 lM Km; kcat/Km = 0.57 lM                         [87]
                                            FXI                                     –                                                    [88]
                                            Complement C1                           –                                                    [89]
                                            FVII                                    –                                                    [90]
                                            Plasminogen                             –                                                    [91]
                                            High-Mr kininogen                       –                                                    [89]
b-FXIIa                                     Prekallikrein                           2.1 lM Km; kcat/Km = 1.67 lM                         [87]
Autoactivation of FXII                      FXII                                    2.4 lM Km; kcat/Km = 0.02 lM                         [92]
Plasma kallikrein                           FXII                                    11 lM Km; kcat/Km = 0.57 lM                          [88]
                                            Single-chain urokinase                  0.064 lM Km                                          [22]
                                            High-Mr kininogen                       1.4 lM Km; kcat/Km = 0.46 lM                         [93]
Prolylcarboxypeptidase                      Prekallikrein                           0.007 lM Km                                          [35]



Table 2 Inhibitors of the enzymes of the plasma kallikrein–kinin system   independently of HK [22–24]. FXI also binds to prothrombin
Enzyme        Inhibitor             Inhibition constant+     Reference    and the glycoprotein Iba–IX–V complex on platelets [24,25].
                                                                          Membrane-binding proteins of HK include gC1qR, urokinase
a-Factor      C1 inhibitor          222.0 · 103 M)1 min)1    [9]          plasminogen activator receptor (u-PAR), and cytokeratin 1
 (F) XIIa     a2-Antiplasmin        11.0 · 103 M)1 min)1     [9]
                                                                          (CK1) (Fig. 1) [26–29]. When HK is proteolyzed by plasma
              a2-Macroglobulin      5.3 · 103 M)1 min)1      [9]
              Antithrombin          1.3 · 103 M)1 min)1      [9]          kallikrein or other proteases to form cleaved HK (HKa),
Plasma        C1 inhibitor          102.0 · 104 M)1 min)1    [9]          membrane tropomysin also functions as a binding site uniquely
 kallikrein   a2-Macroglobulin      69.0 · 104 M)1 min)1     [10]         for this form of kininogen [30]. FXII also has been shown to
              Antithrombin          1.8 · 104 M)1 min)1      [10]         bind to gC1qR, u-PAR, and CK1 [27,31]. Both PK and FXI
              a1-Antitrypsin        0.025 · 104 M)1 min)1    [10]
                                                                          circulate in plasma almost completely bound to HK, but PK
              PAI-1                 360.0 · 104 M)1 min)1    [94]
              Protein C inhibitor   600.0 · 104 M)1 min)1    [95]         binding to endothelial cells predominates [24]. The reasons for
                                                                          this are as follows: (i) the concentration of PK (450 nM) is more
PAI-1, plasminogen activator inhibitor-1.
+                                                                         than tenfold greater than that of FXI (30 nM) in plasma; and
 The values are second-order rate constants.
                                                                          (ii) the free Zn2+ concentration required for PK binding is only
                                                                          0.3 lM, whereas that for FXI binding is 7 lM [24].
ordering of the adsorbed protein molecules that lead to                       When HK and PK assemble on endothelial cells and matrix,
expression of its active site [13]. Negatively charged surfaces           plasma kallikrein activity arises independently of added FXIIa.
consist of artificial materials as found in coagulation assays such        This event occurs in the presence of neutralizing antibody to
as kaolin, celite, and glass surfaces. Several physiologic
substances, such as articular cartilage, skin, fatty acids, endo-
toxin, sodium urate crystals, calcium pyrophosphate, L-homo-
cysteine, hematin, protoporphyrin, sulfatides, heparins,
chondrotin sulfates, and amyloid b-protein, also support
autoactivation of FXII. Formation of activated FXII by
autoactivation results in PK activation with reciprocal activa-
tion of FXII and PK and activation amplification of the system.
In vivo, FXII autoactivation occurs on developing thrombus,
contributing to its extent [7]. Substances that contribute to
Ôcontact activationÕ on a developing thrombus include RNA
from degrading cells, polysomes from platelet membranes, and
fibrin itself [14,15]. FXII activation also occurs under conditions
of sepsis, where bacteria provide a negatively charged surface,
proteases to activate FXII, or a binding site [16,17].
                                                                          Fig. 1. Physiologic assembly and activation of the plasma kallikrein–kinin
                                                                          system. The high molecular weight kininogen (HK)–prekallikrein (PK)
Constitutive activation of the plasma KKS in the                          complex binds to its HUVEC receptor complex, which includes cytoker-
                                                                          atin 1 (CK1), urokinase plasminogen activator receptor (u-PAR) and
intravascular compartment                                                 gC1qR. Prolylcarboxypeptidase (PRCP) bound to the complex activates
It has been recognized that HK, FXII and PK specifically,                  PK to form plasma kallikrein (KAL). The KAL cleaves HK and acti-
                                                                          vates FXII and single-chain urokinase plasminogen activator (Scu-PA).
saturably and reversibly bind to endothelial cells, platelets             Cleaved HK liberates bradykinin (BK), which is a potent activator of
and granulocytes [18–21]. HK serves as the major binding site             tissue-type plasminogen activator (t-PA), NO (nitric oxide) and prosta-
for PK and FXI, although both bind to endothelial cells                   cyclin (PGI2) liberation from endothelial cells. HKa, cleaved HK.


                                                                                 Ó 2007 International Society on Thrombosis and Haemostasis
The changing kallikrein–kinin system 2325

FXII and FXII-deficient plasma, but not PK-deficient plasma
                                                                    Vascular activities of the plasma KKS
[22,32]. The plasma kallikrein formed results in kinetically
favorable single-chain urokinase activation (Km = 64 nM)            Regulation of blood pressure and flow Local BK formation
(Table 1) [22]. The plasma kallikrein on endothelial cells also     is known to influence blood pressure. BK is liberated from HK
results in kinetically favorable FXII activation [33]. These data   by plasma or tissue kallikrein cleavage. The nine amino acid
provide an alternative hypothesis to contact activation for         BK peptide, RPPGFSPFR, has two intravascular receptors:
FXIIa formation in vivo. The increased requirements for free        B2R, which is constitutively expressed, and the BK B1 receptor
Zn2+ for FXII binding to endothelial cells suggest that FXIIÕs      (B1R), which becomes expressed in inflammatory states. BK
association and activation on endothelial cells follows HK and      binds to B2R, a seven-transmembrane G-protein-coupled
PK assembly and activation [24,31]. This proposed mechanism         receptor, and stimulates its G-proteins to release nitric oxide
for PK activation in vivo may be occurring constitutively.          (NO), prostaglandin I2 (prostacyclin), smooth muscle
Firstly, C1INH knockout mice have constitutive tissue edema         hyperpolarization factor, and superoxide [43–46]. In sepsis,
due to increased BK, as it is blocked by a B2R antagonist or by     excessive BK release contributes to hypotension.
mating C1INH and B2R knockout mice [11]. As plasma BK                   BK produced by the plasma and tissue KKS influences
only arises from plasma kallikrein formation and C1INH only         cardiovascular physiology. B2R knockout mice are not consti-
inhibits plasma kallikrein, not tissue kallikein, BK must be        tutively hypertensive; however, upon being subjected to a salt
constantly formed in vivo to give the paw edema seen [11].          load, they have early-onset salt-sensitive hypertension [47]. B2R
Secondly, FXII knockout mice also have plasma BK formation          is involved in the control of regional vascular tone in the coro-
without the presence of FXII [34].                                  nary arteries and the kidneys. The cardioprotective effects of
   A PK activator was purified from endothelial cells [35]. On       angiotensin-converting enzyme (ACE) inhibition, which inhibits
amino acid sequencing, it was identified as the serine protease      BK degradation, is lost in B2R knockout mice. In diabetic mice,
prolylcarboxypeptidase (PRCP) [35]. The Km of PRCP activa-          the absence of B2R increases oxidative stress, mitochondrial
tion of plasma PK (Km = 7 nM) is two hundred and fifty- to           DNA damage, and senescence-associated phenotypes [48]. In
three hundredfold higher than that for activated forms of FXII      tissue kallikrein knockout mice, with reduced tissue BK
(Table 1). This suggests that PRCP activation of PK is favored      formation, there is thinning of the septum and posterior wall
over that of a-FXIIa or b-FXIIa in vivo (Table 1). It is of note    of the heart, resulting in ventricular dilatation and reduced left
that C1INH is a tighter inhibitor of plasma kallikrein than of      ventricular mass [49]. Furthermore, genetic kininogen deficiency
activated FXII, suggesting that plasma kallikrein regulation is     in rats contributes to aortic aneurysm formation [50].
more important than that of FXIIa (Table 2). PRCP was first
recognized as a degrading enzyme for BK and angiotensin II          Thrombosis risk Emerging information indicates that the
(Ki  1 and 0.15 mM, respectively) by cleaving Pro-X bonds on       plasma KKS influences thrombosis risk independently of
the C-terminus of the protein [36]. Both purified and                hemostasis [7,8]. Patients with FXII, PK and HK deficiency are
recombinant PRCP activate PK with a Km  7–17 nM                    exceedingly rare, and although they do not bleed, there are too
[35,37]. Although thought to be lysosomal in origin, PRCP is        few patients to characterize a common clinical phenotype.
a membrane and matrix protein, as it can be demonstrated to be      FXII deficiency is more common than HK or PK deficiency.
there functionally and immunochemically and it was interrupted      Clinical investigations for venous thrombosis risk or on
by a gene trap targeted to membrane proteins [35,37–39]. PRCP       polymorphisms of FXII and their influence on cardiovascular
is a risk factor for metabolic syndrome in men, and a PRCP          disease have been conflicting (see below). The clearest
polymorphism is associated with pre-eclampsia in women              information on thrombosis risk or risk amelioration has been
[40,41]. CHO cells with overexpressed PRCP have increased           derived from animal models, which demonstrate unexpected
PK-activating activity over controls; treatment of these cells      findings.
with small interfering RNA reduces the PK activation on these
cells [42]. Finally, transfected CHO cells mostly express PRCP      BK and kininogen BK infusion is a potent stimulant for tissue-
on their membranes. These combined studies indicate that there      type plasminogen activator (t-PA) release in rabbits and humans
is a constitutive, physiologic endothelial cell mechanism for PK    [51]. Kininogen itself has been shown to have antithrombin
activation independent of FXII autoactivation by contact.           activities. Both HK and low molecular weight kininogen at 5%
                                                                    of their physiologic concentrations block thrombin-induced
                                                                    platelet aggregation and serotonin release by inhibiting
Activities of the plasma KKS
                                                                    thrombin binding to platelets [52]. The thrombin inhibitory
The studies described above reveal a means for KKS assembly         regions of kininogen have been associated with domains 3 and 4,
and activation by physiologic and pathophysiologic mecha-           the BK region [53,54]. A peptide comprising the first five amino
nisms. Several vascular and cellular activities derive from these   acids of BK, RPPGF, was found to bind weakly to the active site
pathways. KKS vascular activities include regulation of blood       of thrombin upon cocrystallization, and to bind the exodomains
pressure and flow and thrombosis risk; the cellular activities       of protease-activated receptor (PAR)1 and PAR4 to prevent
include cellular proliferation, growth, angiogenesis, apoptosis,    thrombin cleavage [55,56]. RPPGF inhibits in vitro and, when
and inflammation.                                                    infused in dogs and humans, ex vivo thrombin-induced platelet

Ó 2007 International Society on Thrombosis and Haemostasis
2326 A. H. Schmaier and K. R. McCrae

aggregation [57,58]. RPPGF in pharmacologic doses prevents                   binds to the overexpressed angiotensin receptor 2 to increase
carotid artery thrombosis in mice and coronary artery                        NO and prostacyclin, and prolong the bleeding time of the
thrombosis in dogs [57,59,60].                                               animal [8] (Fig. 2). Thirdly, RPPGF is elevated in these animals,
   As BK induces NO, prostacyclin and t-PA release from                      due to increased BK degradation by ACE [8]. The elevation of
endothelial cells, we hypothesized that the B2R knockout mouse               RPPGF levels may also contribute to the thrombosis protection.
would be prothrombotic. To our surprise, B2R knockout mice                   These combined studies indicate that BK and its receptor system
have delayed carotid artery occlusion times in the Rose Bengal               indirectly influence thrombosis risk by influencing endothelial
model (Fig. 2) [8]. The mechanism for thrombosis protection is               cell biology through cross-talk with components of the plasma
dependent on this systemÕs interaction with the renin–angioten-              RAS. Such a pathway for risk modification of intra-arterial
sin system (RAS) [61]. In the RAS, angiotensinogen is converted              thrombosis has not been previously appreciated.
to angiotensin I by renin and then converted to angiotensin II by
ACE. ACE also is the major enzyme that degrades BK to BK                     FXII There are conflicts between human clinical and
1–5 (RPPGF) in the intravascular compartment (Fig. 2).                       experimental animal data for the role of FXII in thrombosis
Angiotensin II usually binds to angiotensin receptor 1 to induce             risk. A polymorphism in FXII (46C/T) is associated with
vasoconstriction and salt retention, and elevate blood pressure.             increased risk for arterial thrombosis [63–65]. Individuals
However, if angiotensin receptor 2 is overexpressed, angiotensin             homozygous for the 46C/T polymorphism have lowered FXII
II will preferentially bind to it to induce vasodilatation and               and FXIIa levels. Reduced activated forms of FXII may be
blood pressure reduction. The mechanism by which the B2R                     associated with reduced total fibrinolytic activity, resulting in
knockout mice are protected from thrombosis is 3-fold. Firstly,              increased thrombosis risk. This interpretation is opposite to
in the absence of B2R, angiotensin receptor 2 is overexpressed               what is demonstrated in FXII-deficient mice [7]. FXII-deficient
(Fig. 2). B2R and angiotensin receptor 2 colocalize in cells, and            mice have reduced thrombus after induction of arterial clots
there is an as yet unrecognized mechanism whereby the presence               [7,66]. The mechanism for the increased size of thrombus in
of one GPCR receptor regulates the expression of the other                   mice that have normal levels of FXII may be related to
[8,62]. Secondly, there is increased angiotensin II as a result of           increased contact activation occurring on a developing platelet
reduced BK uptake into cells with reflexive increased ACE                     thrombus [14,15]. Therapeutic inhibition of FXII may result in
degradative activity [8] (Fig. 2). The increased angiotensin II              reduced thrombus formation without bleeding. These
                                                                             observations were not predicted by in vitro investigations on
                                                                             the biochemistry and cell biology of FXII and clinical studies
                                                                             on populations with polymorphisms or defects in FXII.


                                                                             Cellular activities of the plasma KKS
                                                                             Cell proliferation and angiogenesis Investigations have
                                                                             shown that kininogen and related proteins influence cellular
                                                                             activities of endothelial and other cells. These investigations
                                                                             were prompted by the observation that HKa induces selective
                                                                             apoptosis of proliferating endothelial cells and inhibits
                                                                             angiogenesis [67,68]. HKa inhibits neovascularization of s.c.
                                                                             planted Matrigel plugs, as well as fibroblast growth factor
                                                                             2-induced angiogenesis in the chick chorioallantoic membrane
Fig. 2. Mechanisms for thrombosis protection in bradykinin B2 receptor       assay [67,68]. Moreover, peptides from domain 5 of HK (D5),
(B2R) knockout mice. In the absence of B2R, there is increased plasma        which subsumes the HK cell-binding region, induce endothelial
bradykinin, as B2R accounts for 40% of the metabolism of bradykinin.
                                                                             cell apoptosis, inhibit angiogenesis, and are antibacterial
Increased bradykinin results in increased conversion to bradykinin 1–5
(peptide RPPGF) (Blood 2006; 108: 192–99). As a byproduct of increased       [69–71]. Kininogen-deficient Brown Norway Katholiek rats,
RPPGF formation, there are increased levels of angiotensin II (Blood         alternatively, display decreased angiogenesis, possibly resulting
2006; 108: 192–99). Angiotensin-converting enzyme (ACE) also converts        from deficient BK release that is ameliorated by a BK analog or
angiotensin I to angiotensin II. In the absence of B2R, there is increased   kininogen replacement [72,73]. The mechanism(s) by which
expression of the angiotensin receptor 2 (AT2R). The increased angio-
                                                                             these activities occur is not known, but may involve the anti-
tensin II is shunted to overexpressed AT2R, as angiotensin II has the same
binding affinity for angiotensin receptor 1 and AT2R. This leads to a          adhesive function of HKa towards cells on vitronectin, the
paradoxical effect in comparison to the usual angiotensin II elevation.       kininogen multiprotein receptor complex, or tropomyosin
Increased stimulation of AT2R produces vasodilatation and increased          [30,74,75].
plasma nitric oxide (NO) and prostacyclin (PGI2) (Blood 2006; 108: 192–
99). The increased NO and PGI2 prolong the bleeding time, and these          Outside-in signaling mediated by the KKS Although the
animals have delayed thrombosis risk on the Rose Bengal model for
carotid artery thrombosis. These investigations indicate that thrombosis     proangiogenic activities of the KKS are mediated by B1R and
risk can be modified by factors independent of coagulation, fibrinolytic or    B2R, a different receptor system(s) may be involved in the
anticoagulant proteins.                                                      inhibition of cell proliferation, adhesion, anti-apoptosis and

                                                                                   Ó 2007 International Society on Thrombosis and Haemostasis
The changing kallikrein–kinin system 2327

angiogenesis. u-PAR has been shown to mediate intracellular                  4 Ratnoff OD, Davie EW. Waterfall sequence for intrinsic blood clot-
signaling. u-PA binding to u-PAR triggers a cascade of                         ting. Science 1964; 145: 1310–2.
                                                                             5 Weuppers KD, Cochrane CG. Plasma prekallikrein:isolation, charac-
intracellular tyrosine phosphorylation that includes src-type
                                                                               terization, and mechanism of action. J Exp Med 1972; 135: 1–20.
protein kinases, focal adhesion proteins, p38, and extracellular             6 Colman RW, Bagdasarian A, Talamos RC, Seavey M, Scott CF,
signal-related kinase 1/2 (ERK1/2) [76–78]. When HKa or D5                     Kaplan AP. Williams trait. Human kininogen deficiency with dimin-
peptides bind to cultured endothelial cells after basic fibroblast              ished levels of plasminogen proactivator and prekallikrein associated
growth factor-induced cell proliferation, there is a reduction of              with abnormalities of the Hageman factor-dependent pathways. J Clin
                                                                               Invest 1975; 56: 1650–62.
cyclin D1 expression and an upregulation of Cdc2 and cyclin A
                                                                             7 Renne T, Pozgajova M, Gruner S, Schuh K, Pauer HU, Burfeind P,
[79,80]. HKa also inhibits adhesion of endothelial cells to                    Gailani D, Nieswandt B. Defective thrombus formation in mice
vitronectin (90%) and gelatin (40%) without any effect on                      lacking coagulation factor XII. J Exp Med 2005; 280: 28572–80.
adhesion to fibronectin, on which it induces endothelial cell                 8 Shariat-Madar Z, Mahdi F, Warnock M, Homeister JW, Srikanth S,
apoptosis. Endothelial cell migration induced by sphingosine                   Krijanovski Y, Murphey LJ, Jaffa AA, Schmaier AH. Bradykinin B2
                                                                               receptor knockout mice are protected from thrombosis by increased
1-phosphate and vascular endothelial growth factor is blocked
                                                                               nitric oxide and prostacyclin. Blood 2006; 108: 192–9.
by HKa and D5 and may be associated with inhibition of                       9 Pixley RA, Schapira M, Colman RW. The regulation of human
phosphorylation of phosphinositide 3 (PI3)-kinase-Akt and                      factor XIIa by plasma proteinase inhibitors. J Biol Chem 1985; 260:
glycogen synthase kinase (GSK)-3a [81].                                        1723–9.
   u-PAR is known to upregulate Mac-1 adhesion to fibrinogen,                10 Schapira M, Scott CF, Colman RW. Protection of human plasma
                                                                               kallikrein from inactivation by C1 inhibitor and other proteases. The
and focal adhesion kinase (FAK) and mitogen-activated protein
                                                                               role of high molecular weight kininogen. Biochemistry 1981; 20: 2738–
kinase are involved in this process [82]. Furthermore, u-PAR                   43.
binds vitronectin and HKa, and D5 disrupts u-PAR–integrin                   11 Han ED, MacFarlane RC, Mulligan AN, Scafidi J, Davis AE III.
and u-PAR–vitronectin interactions [74,83]. FXII activates                     Increased vascular permeability in C1 inhibitor-deficient mice medi-
ERK1/2 in HEPG2 cells and cultured vascular smooth muscle                      ated by the bradykinin type 2 receptor. J Clin Invest 2002; 109: 1057–
                                                                               63.
cells [84]. Vitronectin binds to the same region on u-PAR as HK,
                                                                            12 Wiggins RC, Cochran CG. The autoactivation of rabbit Hageman
HKa, single-chain urokinase plasminogen activator (Scu-PA),                    factor. J Exp Med 1979; 150: 1122–33.
and FXII (unpublished) [75,85,86]. Preliminary studies indicate             13 Chen X, Wang J, Paszti Z, Wang F, Schrauben JN, Tarabara VV,
that Scu-PA or FXII upregulate ERK1/2 and Akt on cultured                      Schmaier AH, Chen Z. Ordered adsorption of coagulation factor XII
endothelial cells, whereas HKa and D5-derived peptides as well                 on negatively charged polymer surfaces probed by sum frequency gen-
                                                                               eration vibrational spectroscopy. Anal Bioanal Chem 2007; 388: 65–72.
as peptides from domain 2 of u-PAR block this interaction [86].
                                                                            14 Kannemeier C, Shibamiya A, Nakazawa F, Trusheim H, Ruppert C,
The upregulation of ERK1/2 and Akt is mediated by a                            Markart P, Song Y, Tzima E, Kennerknecht E, Niepmann M, von
b1-integrin, is independent of lipid rafts, and is associated with             Bruehl ML, Sedding D, Massberg S, Gunther A, Engelmann B,
increased endothelial cell proliferation and incorporation of                  Preissner KT. Extracellular RNA constitutes a natural procoagulant
5-bromo-2¢-deoxy-uridine [86]. A u-PAR-mediated signaling                      cofactor in blood coagulation. Proc Natl Acad Sci USA 2007; 104:
                                                                               6388–93.
system may be the additional pathway leading to cell prolifer-
                                                                            15 Smith SA, Mutch NJ, Baskar D, Rohloff P, Docampo R, Morrissey
ation, growth and, perhaps, apoptosis and angiogenesis                         JH. Polyphosphate modulates blood coagulation and fibrinolysis. Proc
modulated by HK and its cleavage products.                                     Natl Acad Sci USA 2006; 103: 903–8.
                                                                            16 Matsumoto K, Yamamoto T, Kamata R, Maeda H. Pathogenesis of
                                                                               serratial infection: activation of the Hageman–prekallikrein cascade by
Acknowledgements                                                               serratial protease. J Biochem 1984; 96: 739–49.
                                                                            17 Herwald H, Morgelin M, Olsen A, Rhen M, Dahlback B, Muller-
This work was supported in part by grants HL052779,                            Esterl W. Activation of the contact-phase system on bacterial surfaces
HL055709 and HL086038 to A. H. Schmaier, and grants                            is a clue to serious complications in factious disease. Nat Med 1998; 4:
HL076810, CA83134 and P50HL081011 to K. R. McCrae.                             298–302.
                                                                            18 Gustafson EG, Schutsky D, Knight L, Schmaier AH. High molecular
                                                                               weight kininogen binds to unstimulated platelets. J Clin Invest 1986;
Disclosure of Conflict of Interests                                             78: 310–8.
                                                                            19 Gustafson EJ, Schmaier AH, Wachtfogel YT, Kaufman N, Kucich U,
The authors state that they have no conflict of interest.                       Colman RW. Human neutrophils contain and bind high molecular
                                                                               weight kininogen. J Clin Invest 1989; 84: 28–25.
                                                                            20 Schmaier AH, Kuo A, Lundberg D, Murray SC, Cines DB. Expres-
References                                                                     sion of high molecular weight kininogen on human umbilical vein
                                                                               endothelial cells. J Biol Chem 1988; 263: 16327–33.
 1 Sainz IM, Pixley RA, Colman RA. Fifty years of research on the
                                                                            21 Reddigari SR, Shibayama Y, Brunnee T, Kaplan AP. Human Hag-
   plasma kallikrein–kinin system: from protein structure and function to
                                                                               eman factor (factor XII) and high molecular weight kininogen compete
   cell biology and in-vivo pathophysiology. Thromb Haemost 2007; 98:
                                                                               for the same binding site on human umbilical vein endothelial cells.
   77–83.
                                                                               J Biol Chem 1993; 268: 11982–7.
 2 Gailani D, Renne T. The intrinsic pathway of coagulation: a target for
                                                                            22 Motta G, Rojkjaer R, Hasan AAK, Cines DB, Schmaier AH. High
   treating thromboembolic disease. J Thromb Haemost 2007; 5: 1106–12.
                                                                               molecular weight kininogen regulates prekallikrein assembly and
 3 Ratnoff OD, Colopy JE. A familial hemorrhagic trait associated with a
                                                                               activation on endothelial cells: a novel mechanism for contact activa-
   deficiency of a clot promoting fraction of plasma. J Clin Invest 1955;
                                                                               tion. Blood 1998; 91: 516–28.
   34: 602–13.


Ó 2007 International Society on Thrombosis and Haemostasis
2328 A. H. Schmaier and K. R. McCrae

23 Shariat-Madar Z, Mahdi F, Schmaier AH. Factor XI assembly and              42 Shariat-Madar Z, Rahimi E, Mahdi F, Schmaier AH. Over-expression
   activation on human umbilical vein endothelial cells in culture. Thromb       of prolylcarboxypeptidase enhances plasma prekallikrein activation on
   Haemost 2001; 85: 544–51.                                                     Chinese hamster ovary cells. Am J Physiol Heart Circ Physiol 2005;
24 Mahdi F, Shariat-Madar Z, Schmaier AH. The relative priority of               289: H2697–703.
   prekallikrein and factors XI/XIa assembly on cultured endothelial          43 Palmer RMJ, Ferrige AG, Moncada S. Nitric oxide release accounts
   cells. J Biol Chem 2003; 278: 43983–90.                                       for the biologic activity of endothelium-derived relaxing factor. Nature
25 Yun TH, Baglia FA, Myles T, Navaneetham D, Lopez JA, Walsh PN,                1987; 372: 524–6.
   Leung LL. Thrombin activation of factor XI on activated platelets          44 Hong SL. Effect of bradykinin and thrombin on prostacyclin synthesis
   requires the interaction of factor XI and platelet glycoprotein Ib alpha      in endothelial cells from calf and pig aorta and human umbilical cord
   with thrombin anion-binding exosites I and II, respectively. J Biol           vein. Thromb Res 1980; 18: 787–95.
   Chem 2003; 278: 48112–9.                                                   45 Feletou M, Vanhoutte PM. Endothelium-derived hyperpolarization
26 Herwald H, Dedio J, Kellner R, Loos M, Muller-Esterl W. Isolation             factor: where are we now? Arterioscler Thromb Vasc Biol 2006; 26:
   and characterization of the kininogen binding protein p33 from                1215–25.
   endothelial cells. J Biol Chem 1996; 271: 13040–7.                         46 Holland JA, Pritchard KA, Pappolla MA. Bradykinin induces
27 Joseph K, Ghebrehiwet B, Peerschke EIB, Reid KBM, Kaplan AP.                  superoxide anion release from human endothelial cells. J Cell Physiol
   Identification of the zinc-dependent endothelial cell binding protein for      1990; 143: 21–5.
   high molecular weight kininogen and factor XII: identity with the          47 Cervenka L, Harrison-Bernard LM, Dipp S, Primrose G, Imig JD, El-
   receptor that binds to the globular ÔheadsÕ of C1q (qC1qR). Proc Natl         Dahr SS. Early onset salt-sensitive hypertension in bradykinin B2
   Acad Sci USA 1996; 93: 8552–7.                                                receptor null mice. Hypertension 1999; 34: 176–80.
28 Colman RW, Pixley RA, Najamunnisa S, Yan W, Wang J, Mazar A,               48 Kakoki M, Kizer CM, Yi X, Takahashi N, Kim H-S, Bagnell CR,
   McCrae KR. Binding of high molecular weight kininogen to human                Edgell C-J, Maeda N, Jennette JC, Smithies O. Senescence-associated
   endothelial cells is mediated via a site within domains 2 and 3 of the        phenotypes in Akita diabetic mice are enhanced by absence of
   urokinase receptor. J Clin Invest 1997; 100: 1481–7.                          bradykinin B2 receptors. J Clin Invest 2006; 116: 1302–9.
29 Hasan AAK, Zisman T, Schmaier AH. Identification of cytokeratin 1           49 Meneton P, Bloch-Faure M, Hagege AA, Ruetten H, Huang W,
   as a binding protein and presentation receptor for kininogens on              Bergaya S, Ceiler D, Gehring D, Martins I, Salmon G, Boulanger C,
   endothelial cells. Proc Natl Acad Sci USA 1998; 95: 3615–20.                  Nussberger J, Crozatier B, Gasc J-M, Heudes D, Bruneval P,
30 Zhang J-C, Donate F, Qi X, Ziats NP, Juarez JC, Mazar AP, Pang Y-             Doetschman T, Menard J, Alhenc-Gelas F. Cardiovascular
   P, McCrae KR. The antiangiogenic activity of cleaved high molecular           abnormalities with normal blood pressure in tissue kallikrein-deficient
   weight kininogen is mediated through binding to endothelial cell              mice. Proc Natl Acad Sci USA 2001; 98: 2634–9.
   tropomyosin. Proc Natl Acad Sci USA 2002; 99: 12224–9.                     50 Kaschina E, Stoll M, Sommerfeld M, Steckelings UM, Kreutz R,
31 Mahdi F, Shariat-Madar Z, Figueroa CD, Schmaier AH. Factor XII                Unger T. Genetic kininogen deficiency contributes to aortic aneu-
   interacts with the multiprotein assembly of urokinase plasminogen             rysm formation but not to atherosclerosis. Physiol Genomics 2004; 19:
   activator receptor, gC1qR, and cytokeratin on endothelial cell mem-           41–9.
   branes. Blood 2002; 99: 3585–96.                                           51 Brown NJ, Gainer JV, Stein CM, Vaughan DE. Bradykinin stimulates
32 Motta G, Shariat-Madar Z, Mahdi F, Sampaio CAM, Schmaier AH.                  tissue plasminogen activator release in human vasculature. Hyperten-
   Assembly and activation of high molecular weight kininogen and                sion 1999; 33: 1431–5.
   prekallikrein on cell matrix. Thromb Haemost 2001; 86: 840–7.              52 Meloni FJ, Schmaier AH. Low molecular weight kininogen binds to
33 Rojkjaer R, Hasan AAK, Motta G, Schousboe I, Schmaier AH.                     platelets to modulate thrombin-induced platelet activation. J Biol
   Factor XII does not initiate prekallikrein activation on endothelial          Chem 1991; 266: 6786–94.
   cells. Thromb Haemost 1998; 80: 74–81.                                     53 Jiang Y, Muller-Esterl W, Schmaier AH. Domain 3 of kininogens
34 Iwaki T, Castellino FJ. Plasma levels of bradykinin are suppressed in         contains a cell binding site and a site that modifies thrombin activation
   factor XII-deficient mice. Thromb Haemost 2006; 95: 1003–10.                   of platelets. J Biol Chem 1992; 267: 3712–7.
35 Shariat-Madar Z, Mahdi F, Schmaier AH. Identification and char-             54 Hasan AAK, Amenta S, Schmaier AH. Bradykinin and its metabolite
   acterization of prolylcarboxypeptidase as an endothelial cell prekal-         ARG-PRO-PRO-GLY-PHE are selective inhibitors of a-thrombin-
   likrein activator. J Biol Chem 2002; 277: 17962–9.                            induced platelet activation. Circulation 1996; 94: 517–28.
36 Oyda CE, Marinkovic DV, Hammon KJ, Stewart TA, Erdos EG.                   55 Hasan AAK, Warnock M, Nieman M, Srikanth S, Mahdi F, Krish-
   Purification and properties of prolylcarboxypeptidase (angiotensinase          nan R, Tulinsky A, Schmaier AH. The mechanisms of Arg-Pro-Pro-
   C) from human kidney. J Biol Chem 1978; 253: 5927–31.                         Gly-Phe inhibition of thrombin. Amer J Physiol Heart and Circ Physiol
37 Shariat-Madar Z, Mahdi F, Schmaier AH. Recombinant prolyl-                    2003; 285: H183–93.
   carboxypeptidase activates plasma prekallikrein. Blood 2004; 103:          56 Nieman MT, Pagan-Ramos E, Warnock M, Krijanovski Y, Hasan
   4554–61.                                                                      AAK, Schmaier AH. Mapping the interaction of bradykinin 1–5 with
38 Moreira CR, Schmaier AH, Mahdi F, da Motta G, Nader HB, Sha-                  the exodomain of protease activated receptor 4 (PAR4). FEBS Lett
   riat-Madar Z. Identification of prolylcarboxypeptidase as the cell             2005; 579: 25–9.
   matrix-associated prekallikrein activator. FEBS Lett 2002; 523: 167–       57 Hasan AAK, Rebello SS, Smith E, Srikanth S, Werns S, Driscoll E,
   70.                                                                           Faul J, Brenner D, Normolle D, Lucchesi BR, Schmaier AH.
39 Skarnes WC. Gene trapping methods for the identification and func-             Thrombostatin inhibits induced canine coronary thrombosis. Thromb
   tional analysis of cell surface proteins in mice. Methods Enzymol 2000;       Haemost 1999; 82: 1182–7.
   328: 592–615.                                                              58 Murphey LJ, Malave HA, Petro J, Biaggioni I, Byrne DW, Vaughan
40 McCarthy JJ, Meyer J, Moliterno DJ, Newby LK, Rogers WJ, Topol                DE, Luther JM, Pretorius M, Brown NJ. Bradykinin and its metab-
   EJ, GenQuest multicenter study. Evidence for substantial effect mod-           olite bradykinin 1–5 inhibit thrombin-induced platelet aggregation in
   ification by gender in a large scale genetic association study of the          humans. J Pharm Exp Ther 2006; 318: 1287–92.
   metabolic syndrome among coronary heart disease patients. Hum              59 Nieman MT, Warnock M, Hasan AAK, Mahdi F, Lucchesi BR,
   Genet 2003; 114: 87–98.                                                       Brown NJ, Murphey LJ, Schmaier AH. The preparation and char-
41 Wang L, Feng Y, Zhang Y, Zhou H, Jiang S, Niu T, Wei LJ, Xu X, Xu             acterization of novel peptide antagonists to thrombin, factor VIIa and
   X, Wang X. Prolylcarboxypeptidase gene, chronic hypertension, and             activation of protease activates receptor 1. J Pharm Exp Ther 2004;
   risk of preeclampsia. Am J Obstet Gynecol 2006; 195: 162–71.                  311: 492–501.



                                                                                     Ó 2007 International Society on Thrombosis and Haemostasis
The changing kallikrein–kinin system 2329

60 Hasan AAK, Schmaier AH, Warnock M, Normolle D, Driscoll E,                       tion of focal adhesion proteins and activation of mitogen-activated
   Lucchesi BR, Werns SW. Thrombostatin inhibits cyclic flow variations              protein kinase in cultured endothelial cells. J Biol Chem 1998; 273:
   in stenosed canine coronary arteries. Thromb Haemost 2001; 86: 1296–             18268–72.
   304.                                                                        78   Nguyen DHD, Webb DJ, Catling AD, Song Q, Dhakephalkar A,
61 Schmaier AH. The kallikrein–kinin and the rennin–angiotensin sys-                Weber MJ, Ravichandran KS, Gonias SL. Urokinase-type plasmin-
   tems have a multilayered interaction. Am J Physiol Regul Integr Comp             ogen activator stimulates the Ras/Extracellular signal-regulated kinase
   Physiol 2003; 285: R1–13.                                                        (ERK) signaling pathway and MCF-7 cell migration by a mechanism
62 Abadir PM, Periasamy A, Carey RM, Siragy HM. Angiotensin II type                 that requires focal adhesion kinase, Src, and Shc. J Biol Chem 2000;
   2 receptor–bradykinin B2 receptor functional heterodimerization.                 275: 19382–8.
   Hypertension 2006; 48: 316–22.                                              79   Guo Y-L, Wang S, Colman RW. Kininostatin, an angiogenic inhibi-
63 Soria JM, Almasy L, Souto JC, Bacq D, Buil A, Faure A, Martinez-                 tor, inhibits proliferation and induces apoptosis of human endothelial
   Marchan E, Mateo J, Borrell M, Stone W, Lathrop M, Fontcuberta J,                cells. Arterioscler Thromb Vasc Biol 2001; 21: 1427–33.
   Blangero J. A quantitative-trait locus in the human factor XII gene         80   Wang S, Hasham MG, Isordia-Salas I, Tsygankov AY, Colman RW,
   influences both plasma factor XII levels and susceptibility to throm-             Guo Y-L. Upregulation of Cdc2 and cyclin A during apoptosis of
   botic disease. Am J Hum Genet 2002; 70: 567–74.                                  endothelial cells induced by cleaved high-molecular-weight kininogen.
64 Zito F, Lowe GDO, Rumley A, McMahon AD, Humphries SE.                            Am J Physiol Heart Circ Physiol 2003; 284: H1917–23.
   Association of the factor XII 46CT polymorphism with risk of               81   Katkade V, Soyombo AA, Isordia-Salas I, Bradford HN, Gaughan
   coronary heart disease in the WOSCOPS study. Atherosclerosis 2002;               JP, Colman RW, Panetti TS. Domain 5 of cleaved high molecular
   165: 153–8.                                                                      weight kininogen inhibits endothelial cell migration through Akt.
65 Colhoun HM, Zito F, Chan NN, Rubens MB, Fuller JH, Humphries                     Thromb Haemost 2005; 94: 606–14.
   SE. Activated factor XII levels and factor XII 46CT genotype in            82   Zhang H, Colman RW, Sheng N. Regulation of CD11b/CD18 (Mac-
   relation to coronary artery calcification in patients with type 1 diabetes        1) adhesion to fibrinogen by urokinase receptor (uPAR). Inflamm Res
   and healthy subjects. Atherosclerosis 2002; 163: 363–9.                          2003; 52: 86–93.
66 Kleinschnitz C, Stoll G, Bendszuz M, Schuh K, Pauer HU, Burfeind P,         83   Cunningham O, Andolfo A, Santovito ML, Iuzzolino L, Blasi F,
   Renne C, Gailani D, Nieswandt B, Renne T. Targeting coagulation                  Sidenius N. Dimerization controls the lipid raft partitioning of uPAR/
   factor XII provides protection from pathologic thrombosis in cerebral            CD87 and regulates its biologic functions. EMBO J 2003; 22: 5994–
   ischemia without interfering with hemostasis. J Exp Med 2006; 203:               6003.
   513–8.                                                                      84   Gordon EM, Venkatesan N, Salazat R, Tang H, Schmeidler-Sapiro K,
67 Zhang J-C, Claffey K, Sakthivel R, Darzynkiewicz Z, Shaw DE, Leal                 Buckley S, Warburton D, Hall FL. Factor XII-induced mitogenesis is
   J, Wang Y-C, Lu F-M, McCrae KR. Two-chain high molecular weight                  mediated via a distinct signal transduction pathway that activates a
   kininogen induces endothelial cell apoptosis and inhibits angiogenesis:          mitogen-activated protein kinase. Proc Natl Acad Sci USA 1996; 93:
   partial activity within domain 5. FASEB J 2000; 14: 2589–600.                    2174–9.
68 Colman RW, Jameson BA, Lin Y, Johnson D, Mousa SA. Domain 5                 85   Li Y, Lawrence DA, Zhang L. Sequences within domain II of the
   of high molecular weight kininogen (kininostatin) down-regulates                 urokinase receptor critical for differential ligand recognition. J Biol
   endothelial cell proliferation and migration and inhibits angiogenesis.          Chem 2003; 278: 29925–32.
   Blood 2000; 95: 543–50.                                                     86   Schmaier AH, Mahdi F, Sitrin R. The urokinase plasminogen acti-
69 Zhang J-C, Qi X, Juarez J, Plunkett M, Donate F, Sakthivel R, Mazar              vator receptor mediates ScuPA- or FXII-induced cell growth and
   AP, McCrae KR. Inhibition of angiogenesis by two-chain high                      proliferation. Blood 2006; 108(Suppl.): 1817 (abstract).
   molecular weight kininogen (HKa) and kininogen-derived polypep-             87   Tankersley DL, Finlayson JS. Kinetics of activation and autoactiva-
   tides. Can J Physiol Pharmacol 2002; 80: 85–90.                                  tion of human factor XII. Biochemistry 1984; 23: 273–9.
70 Hasan AAK, Cines DB, Herwald H, Schmaier AH, Muller-Esterl W.               88   Kurachi K, Fujikawa K, Davie EW. Mechanism of activation of bo-
   Mapping the cell binding site on high molecular weight kininogenÕs               vine factor XI by factor XII and factor XIIa. Biochemistry 1980; 19:
   domain 5. J Biol Chem 1995; 270: 19256–61.                                       1330–8.
71 Nordahl EA, Rydengard V, Morgelin M, Schmidtchen A. Domain 5                89   Ghrbrehiwet B, Randazzo BP, Dunn JT, Silverberg M, Kaplan AP.
   of high molecular weight kininogen is antibacterial. J Biol Chem 2005;           Mechanisms of activation of the classical pathway of complement by
   280: 34832–9.                                                                    Hageman factor fragment. J Clin Invest 1983; 71: 1450–6.
72 Hu DE, Fan TP. [Leu8]des-Arg9-bradykinin inhibits the angiogenic            90   Seligsohn U, Osterud B, Brown SF, Rappaport SI. Activation of hu-
   effect of bradykinin and interleukin-1 in rats. Br J Pharmacol 1993;              man factor VII in human plasma and purified systems. J Clin Invest
   109: 14–7.                                                                       1979; 64: 239–43.
73 Hayashi I, Amano H, Yoshida S, Kamata K, Kamata M, Inukai M,                91   Goldsmith GH, Saito H, Ratnoff OD. The activation of plasminogen
   Fujita T, Kumagai Y, Furudat S, Majima M. Suppressed angiogenesis                by Hageman factor (Factor XII) and Hageman Factor fragments. J
   in kininogen-deficiencies. Lab Invest 2002; 82: 871–80.                           Clin Invest 1978; 62: 54–60.
74 Chavakis T, Kanse SM, Lupu F, Hammes H-P, Muller-Esterl W,                  92   Bernardo MM, Day DE, Olson ST, Shore JD. Surface-independent
   Pixley RA, Colman RW, Preissner KT. Different mechanisms define                    acceleration of factor XII activation by zinc ions. I Kinetic charac-
   the antiadhesive function of high molecular weight kininogen in inte-            terization of the metal ion rate enhancement. J Biol Chem 1993; 268:
   grin- and urokinase receptor-dependent interactions. Blood 2000; 96:             12468–76.
   514–22.                                                                     93   Tayeh MA, Olson ST, Shore JD. Surface-induced alterations in the
75 Mahdi F, Shariat-Madar Z, Kuo A, Carinato M, Cines DB, Schmaier                  kinetic pathway for cleavage of human high molecular weight kinin-
   AH. Mapping the interaction between high molecular weight kinino-                ogen by plasma kallikrein. J Biol Chem 1994; 269: 16318–25.
   gen and the urokinase plasminogen activator receptor. J Biol Chem           94   Berrettini M, Schleef RR, Espana F, Loskutoff DJ, Griffin JH.
   2004; 279: 16621–8.                                                              Interaction of type 1 plasminogen activator inhibitor with the
76 Konakova M, Hucho F, Schleuning W-D. Downstream targets of                       enzymes of the contact activation system. J Biol Chem 1989; 264:
   urokinase-type plasminogen-activator-mediated signal transduction.               11738–43.
   Eur J Biochem 1998; 253: 421–9.                                             95   Meijers JCM, Kanters DHA, Vlooswijk RRA, van Erp HE, Hessing
77 Tang H, Kerins DM, Hao Q, Inagami T, Vaughan DE. The urokinase-                  M, Bouma BN. Inactivation of human plasma kallikrein and factor
   type plasminogen activator receptor mediates tyrosine phosphoryla-               Xia by protein C inhibitor. Biochemistry 1988; 27: 4231–7.



Ó 2007 International Society on Thrombosis and Haemostasis

Más contenido relacionado

La actualidad más candente

Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human CancerSima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human CancerSima Lev
 
Lecture 3, spring 2014
Lecture 3, spring 2014Lecture 3, spring 2014
Lecture 3, spring 2014Shabab Ali
 
Hemostatsis (Fibrinolysis - Plasminogen)
Hemostatsis (Fibrinolysis - Plasminogen)Hemostatsis (Fibrinolysis - Plasminogen)
Hemostatsis (Fibrinolysis - Plasminogen)Pradeep Singh Narwat
 
L15 b bleeding disorders my lecture (2)
L15 b bleeding disorders my lecture (2)L15 b bleeding disorders my lecture (2)
L15 b bleeding disorders my lecture (2)Bruno Mmassy
 
Charlton2003
Charlton2003Charlton2003
Charlton2003brandi3
 
Review of the coagulation system
Review of the coagulation systemReview of the coagulation system
Review of the coagulation systemderosaMSKCC
 
4 u1.0-b978-1-4160-4224-2..50043-0..docpdf
4 u1.0-b978-1-4160-4224-2..50043-0..docpdf4 u1.0-b978-1-4160-4224-2..50043-0..docpdf
4 u1.0-b978-1-4160-4224-2..50043-0..docpdfLoveis1able Khumpuangdee
 
Hsp70 and Hsp90
Hsp70 and Hsp90 Hsp70 and Hsp90
Hsp70 and Hsp90 Avin Snyder
 
ABC Proteins Statistical Analysis
ABC Proteins Statistical AnalysisABC Proteins Statistical Analysis
ABC Proteins Statistical AnalysisMehul Garg
 
Blood clotting and role of calcium
Blood clotting and role of calciumBlood clotting and role of calcium
Blood clotting and role of calciumFatima Rahat
 
The Blood and Hemostasis and Blood Coagulation
The Blood and Hemostasis and Blood CoagulationThe Blood and Hemostasis and Blood Coagulation
The Blood and Hemostasis and Blood CoagulationAmany Elsayed
 
Lecture 9, fall 2014
Lecture 9, fall 2014Lecture 9, fall 2014
Lecture 9, fall 2014Shabab Ali
 

La actualidad más candente (20)

Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human CancerSima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
 
Normal coagulation
Normal coagulationNormal coagulation
Normal coagulation
 
Coagulation Cascade
Coagulation CascadeCoagulation Cascade
Coagulation Cascade
 
Lecture 3, spring 2014
Lecture 3, spring 2014Lecture 3, spring 2014
Lecture 3, spring 2014
 
Hemostatsis (Fibrinolysis - Plasminogen)
Hemostatsis (Fibrinolysis - Plasminogen)Hemostatsis (Fibrinolysis - Plasminogen)
Hemostatsis (Fibrinolysis - Plasminogen)
 
Clotting factors
Clotting factorsClotting factors
Clotting factors
 
L15 b bleeding disorders my lecture (2)
L15 b bleeding disorders my lecture (2)L15 b bleeding disorders my lecture (2)
L15 b bleeding disorders my lecture (2)
 
Charlton2003
Charlton2003Charlton2003
Charlton2003
 
Review of the coagulation system
Review of the coagulation systemReview of the coagulation system
Review of the coagulation system
 
4 u1.0-b978-1-4160-4224-2..50043-0..docpdf
4 u1.0-b978-1-4160-4224-2..50043-0..docpdf4 u1.0-b978-1-4160-4224-2..50043-0..docpdf
4 u1.0-b978-1-4160-4224-2..50043-0..docpdf
 
SOT Poster Final 2016
SOT Poster Final 2016SOT Poster Final 2016
SOT Poster Final 2016
 
hi
hihi
hi
 
Hemostasis
HemostasisHemostasis
Hemostasis
 
Hsp70 and Hsp90
Hsp70 and Hsp90 Hsp70 and Hsp90
Hsp70 and Hsp90
 
ABC Proteins Statistical Analysis
ABC Proteins Statistical AnalysisABC Proteins Statistical Analysis
ABC Proteins Statistical Analysis
 
Blood clotting and role of calcium
Blood clotting and role of calciumBlood clotting and role of calcium
Blood clotting and role of calcium
 
final disso
final dissofinal disso
final disso
 
The Blood and Hemostasis and Blood Coagulation
The Blood and Hemostasis and Blood CoagulationThe Blood and Hemostasis and Blood Coagulation
The Blood and Hemostasis and Blood Coagulation
 
Lecture 9, fall 2014
Lecture 9, fall 2014Lecture 9, fall 2014
Lecture 9, fall 2014
 
Fibrinolysis
Fibrinolysis Fibrinolysis
Fibrinolysis
 

Destacado (6)

Trypanosoma
TrypanosomaTrypanosoma
Trypanosoma
 
Trypanosomiasis
TrypanosomiasisTrypanosomiasis
Trypanosomiasis
 
Trypanosoma
TrypanosomaTrypanosoma
Trypanosoma
 
Trypanosomiasis
TrypanosomiasisTrypanosomiasis
Trypanosomiasis
 
Coagulation and hemostasis
Coagulation and hemostasisCoagulation and hemostasis
Coagulation and hemostasis
 
Coagulation
CoagulationCoagulation
Coagulation
 

Similar a The plasma kallikrein kinin system its evolution from contact activation

Coagulants and anticoagulants
Coagulants and anticoagulantsCoagulants and anticoagulants
Coagulants and anticoagulantsLikhita Kolli
 
Killing biological cascades
Killing biological cascadesKilling biological cascades
Killing biological cascadesWilson Ong
 
14 infection inflammation
14 infection inflammation14 infection inflammation
14 infection inflammationazmal sarker
 
Introduction of inflammation
Introduction of inflammationIntroduction of inflammation
Introduction of inflammationshivangimistry3
 
Physiology of Coagulation
Physiology of CoagulationPhysiology of Coagulation
Physiology of CoagulationFarah Jafri
 
Seminario biologia mol
Seminario biologia molSeminario biologia mol
Seminario biologia molluismiguelruiz
 
Leukocyte extravasation
Leukocyte extravasationLeukocyte extravasation
Leukocyte extravasationdevadevi666
 
Chemikines&liver2002[1]
Chemikines&liver2002[1]Chemikines&liver2002[1]
Chemikines&liver2002[1]Andrey Levin
 
Dinâmica da secreção de insulina, dm2,obesidade
Dinâmica da secreção de insulina, dm2,obesidadeDinâmica da secreção de insulina, dm2,obesidade
Dinâmica da secreção de insulina, dm2,obesidadeRuy Pantoja
 
Bleeding disorder (pathology).pdf
Bleeding disorder (pathology).pdfBleeding disorder (pathology).pdf
Bleeding disorder (pathology).pdfRajeebLochanKhatua
 
Ckmb interferencia con macro ck
Ckmb interferencia con macro ckCkmb interferencia con macro ck
Ckmb interferencia con macro ckAnafernandaaire
 
Immune system and inflamatory response to cpb(1)
Immune system and inflamatory response to cpb(1)Immune system and inflamatory response to cpb(1)
Immune system and inflamatory response to cpb(1)Manu Jacob
 

Similar a The plasma kallikrein kinin system its evolution from contact activation (20)

Coagulants and anticoagulants
Coagulants and anticoagulantsCoagulants and anticoagulants
Coagulants and anticoagulants
 
Chemokine receptor~
Chemokine  receptor~Chemokine  receptor~
Chemokine receptor~
 
Killing biological cascades
Killing biological cascadesKilling biological cascades
Killing biological cascades
 
Chemokines
ChemokinesChemokines
Chemokines
 
14 infection inflammation
14 infection inflammation14 infection inflammation
14 infection inflammation
 
Management of severe hyperkalemia
Management of severe hyperkalemiaManagement of severe hyperkalemia
Management of severe hyperkalemia
 
Introduction of inflammation
Introduction of inflammationIntroduction of inflammation
Introduction of inflammation
 
Physiology of Coagulation
Physiology of CoagulationPhysiology of Coagulation
Physiology of Coagulation
 
Seminario biologia mol
Seminario biologia molSeminario biologia mol
Seminario biologia mol
 
Egc
EgcEgc
Egc
 
Overview haemostasis 2
Overview haemostasis 2Overview haemostasis 2
Overview haemostasis 2
 
Warfrin Monitering
Warfrin MoniteringWarfrin Monitering
Warfrin Monitering
 
Leukocyte extravasation
Leukocyte extravasationLeukocyte extravasation
Leukocyte extravasation
 
Pathology.pptx
Pathology.pptxPathology.pptx
Pathology.pptx
 
Chemikines&liver2002[1]
Chemikines&liver2002[1]Chemikines&liver2002[1]
Chemikines&liver2002[1]
 
Dinâmica da secreção de insulina, dm2,obesidade
Dinâmica da secreção de insulina, dm2,obesidadeDinâmica da secreção de insulina, dm2,obesidade
Dinâmica da secreção de insulina, dm2,obesidade
 
Bleeding disorder (pathology).pdf
Bleeding disorder (pathology).pdfBleeding disorder (pathology).pdf
Bleeding disorder (pathology).pdf
 
Blood Coagulation Cascade
Blood Coagulation CascadeBlood Coagulation Cascade
Blood Coagulation Cascade
 
Ckmb interferencia con macro ck
Ckmb interferencia con macro ckCkmb interferencia con macro ck
Ckmb interferencia con macro ck
 
Immune system and inflamatory response to cpb(1)
Immune system and inflamatory response to cpb(1)Immune system and inflamatory response to cpb(1)
Immune system and inflamatory response to cpb(1)
 

Más de LAB IDEA

New fundamentals in hemostasis
New fundamentals in hemostasisNew fundamentals in hemostasis
New fundamentals in hemostasisLAB IDEA
 
Hougie 2004-journal of-thrombosis_and_haemostasis
Hougie 2004-journal of-thrombosis_and_haemostasisHougie 2004-journal of-thrombosis_and_haemostasis
Hougie 2004-journal of-thrombosis_and_haemostasisLAB IDEA
 
Sistema de coagulacion
Sistema de coagulacionSistema de coagulacion
Sistema de coagulacionLAB IDEA
 
Clase parasitosis 2017
Clase parasitosis 2017Clase parasitosis 2017
Clase parasitosis 2017LAB IDEA
 
Enzimas musculo esqueleticas 2017
Enzimas musculo esqueleticas 2017Enzimas musculo esqueleticas 2017
Enzimas musculo esqueleticas 2017LAB IDEA
 
Hígado graso
Hígado grasoHígado graso
Hígado grasoLAB IDEA
 
Historical perspective and future direction of coagulation research
Historical perspective and future direction of coagulation researchHistorical perspective and future direction of coagulation research
Historical perspective and future direction of coagulation researchLAB IDEA
 
When should we measure lipoprotein (a)?
When should we measure lipoprotein (a)?When should we measure lipoprotein (a)?
When should we measure lipoprotein (a)?LAB IDEA
 
Metabolic acidosis nature
Metabolic acidosis natureMetabolic acidosis nature
Metabolic acidosis natureLAB IDEA
 
Alcalosis Metabolica
Alcalosis MetabolicaAlcalosis Metabolica
Alcalosis MetabolicaLAB IDEA
 
Equilibrio ácido base
Equilibrio ácido baseEquilibrio ácido base
Equilibrio ácido baseLAB IDEA
 
Acidosis metabolica
Acidosis metabolicaAcidosis metabolica
Acidosis metabolicaLAB IDEA
 
Daño renal agudo
Daño renal agudoDaño renal agudo
Daño renal agudoLAB IDEA
 
LESION RENAL AGUDA
LESION RENAL AGUDALESION RENAL AGUDA
LESION RENAL AGUDALAB IDEA
 
TRASTORNOS DE LA COAGULACION EN HEPATOPATIAS
TRASTORNOS DE LA COAGULACION EN HEPATOPATIASTRASTORNOS DE LA COAGULACION EN HEPATOPATIAS
TRASTORNOS DE LA COAGULACION EN HEPATOPATIASLAB IDEA
 
HEPATITIS POR VIRUS B
HEPATITIS POR VIRUS BHEPATITIS POR VIRUS B
HEPATITIS POR VIRUS BLAB IDEA
 
Fasting is not routinely required for determination of a lipid profile
Fasting is not routinely required for determination of a lipid profileFasting is not routinely required for determination of a lipid profile
Fasting is not routinely required for determination of a lipid profileLAB IDEA
 

Más de LAB IDEA (20)

New fundamentals in hemostasis
New fundamentals in hemostasisNew fundamentals in hemostasis
New fundamentals in hemostasis
 
Hougie 2004-journal of-thrombosis_and_haemostasis
Hougie 2004-journal of-thrombosis_and_haemostasisHougie 2004-journal of-thrombosis_and_haemostasis
Hougie 2004-journal of-thrombosis_and_haemostasis
 
Sistema de coagulacion
Sistema de coagulacionSistema de coagulacion
Sistema de coagulacion
 
Clase parasitosis 2017
Clase parasitosis 2017Clase parasitosis 2017
Clase parasitosis 2017
 
ICTERICIA
ICTERICIAICTERICIA
ICTERICIA
 
ASCITIS
ASCITISASCITIS
ASCITIS
 
Enzimas musculo esqueleticas 2017
Enzimas musculo esqueleticas 2017Enzimas musculo esqueleticas 2017
Enzimas musculo esqueleticas 2017
 
Hígado graso
Hígado grasoHígado graso
Hígado graso
 
Historical perspective and future direction of coagulation research
Historical perspective and future direction of coagulation researchHistorical perspective and future direction of coagulation research
Historical perspective and future direction of coagulation research
 
When should we measure lipoprotein (a)?
When should we measure lipoprotein (a)?When should we measure lipoprotein (a)?
When should we measure lipoprotein (a)?
 
Metabolic acidosis nature
Metabolic acidosis natureMetabolic acidosis nature
Metabolic acidosis nature
 
Alcalosis Metabolica
Alcalosis MetabolicaAlcalosis Metabolica
Alcalosis Metabolica
 
Equilibrio ácido base
Equilibrio ácido baseEquilibrio ácido base
Equilibrio ácido base
 
Acidosis metabolica
Acidosis metabolicaAcidosis metabolica
Acidosis metabolica
 
Daño renal agudo
Daño renal agudoDaño renal agudo
Daño renal agudo
 
LESION RENAL AGUDA
LESION RENAL AGUDALESION RENAL AGUDA
LESION RENAL AGUDA
 
TRASTORNOS DE LA COAGULACION EN HEPATOPATIAS
TRASTORNOS DE LA COAGULACION EN HEPATOPATIASTRASTORNOS DE LA COAGULACION EN HEPATOPATIAS
TRASTORNOS DE LA COAGULACION EN HEPATOPATIAS
 
HEPATITIS POR VIRUS B
HEPATITIS POR VIRUS BHEPATITIS POR VIRUS B
HEPATITIS POR VIRUS B
 
Fasting is not routinely required for determination of a lipid profile
Fasting is not routinely required for determination of a lipid profileFasting is not routinely required for determination of a lipid profile
Fasting is not routinely required for determination of a lipid profile
 
PTI
PTIPTI
PTI
 

The plasma kallikrein kinin system its evolution from contact activation

  • 1. Journal of Thrombosis and Haemostasis, 5: 2323–2329 REVIEW ARTICLE The plasma kallikrein–kinin system: its evolution from contact activation A . H . S C H M A I E R and K . R . M C C R A E Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, OH, USA To cite this article: Schmaier AH, McCrae KR. The plasma kallikrein–kinin system: its evolution from contact activation. J Thromb Haemost 2007; 5: 2323–9. review will outline physiologic activities of the plasma KKS Summary. The plasma kallikrein–kinin system consists of the that are not emphasized in other recent reviews [1,2]. proteins factor XII (FXII), prekallikrein (PK), and high FXII deficiency (Hageman trait) was discovered by Ratnoff molecular weight kininogen. It was first recognized as a sur- and Colopy in an individual who had prolonged blood clotting face-activated coagulation system that is activated when blood times without bleeding [3]. Activation of FXII results in FXI or plasma interacts with artificial surfaces. Although surface- activation, giving rise to the coagulation cascade [4]. As result activated contact activation occurs in vivo in the case of tissue of elucidating non-FXII-deficient etiologies for isolated pro- destruction or a developing thrombus, the physiologic basis for longed activated partial thromboplastin times (APTT), plasma the activation and function of this system has not been PK and HK were discovered [5,6]. These proteins influence the delineated. New investigations indicate that there is a proteo- 200 million surface-activated coagulation tests, APTTs and lytic pathway on cells for PK activation independent of FXII. activated clotting times performed annually in the USA. New This pathway for PK with subsequent FXII activation indicates interest in FXII has arisen since it has been observed that FXII- physiologic activities. These activities include blood pressure deficient mice have reduced thrombus compared to the wild regulation and modulation of thrombosis risk independently of type [7]. Furthermore, bradykinin (BK) B2 receptor (B2R)- hemostasis. Furthermore, they include regulation of endothelial deficient mice also have reduced thrombosis risk [8]. C1 esterase cell proliferation, angiogenesis and apoptosis through a cellular- inhibitor (C1INH), the SERPIN inhibitor of the enzymes of based, outside-in signaling system. The present characteriza- this system, accounts for 90% of inhibition of FXIIa and 50% tions of this system, which incorrectly had been thought to of inhibition of plasma kallikrein [9,10] (Table 2). Although initiate coagulation, represent an evolution of understanding in plasminogen activator inhibitor-1 and protein C inhibitor, this field. mole for mole, may be more potent inhibitors of plasma kallikrein than C1INH, the intravascular concentration of Introduction C1INH is highest, making it the most important (Table 2). C1INH deficiency is the etiology of hereditary angioedema, a Appreciation of the plasma kallikrein–kinin system (KKS) has disorder associated with unregulated BK formation, producing grown. The KKS consists of two zymogens, factor XII (FXII) secondary angioedema in humans and mice [11]. and prekallikrein (PK), and one substrate/cofactor, high molecular weight kininogen (HK) (Table 1). These proteins influence surface-activated in vitro coagulation assays, but Contact activation of FXII and the proteins of the plasma deficiencies are not associated with bleeding. Recent studies KKS indicate activities in vascular biology, including modulation of The three proteins (FXII, PK, and HK) of the plasma KKS are thrombosis risk independently of blood coagulation. This called Ôcontact factorsÕ because, until recently (see next section), there was no known mechanism for their initiation of activation other than FXII autoactivation on surfaces [2,3]. Autoactiva- Correspondence: Alvin H. Schmaier, Case Western Reserve tion is the event where zymogen FXII becomes an enzyme in the University, University Hospital Case Medical Center, Division of presence of a negatively charged surface, a process twentynine- Hematology and Oncology, 10900 Euclid Avenue WRB2-130, Cleveland, OH 44106-7284, USA. fold less efficient than activation by plasma kallikrein [12] Tel.: +1 216 368 1172; fax: +1 216 368 3014; e-mail: (Table 1). The biochemistry of this phenomenon is not under- schmaier@case.edu stood, but recent studies using sum frequency generation vibrational spectroscopy indicate that FXII autoactivation at Received 3 August 2007, accepted 14 September 2007 the molecular level occurs by imposing specific orientation and Ó 2007 International Society on Thrombosis and Haemostasis
  • 2. 2324 A. H. Schmaier and K. R. McCrae Table 1 Enzymes/substrates of the plasma kallikrein–kinin system Enzyme Substrate Kinetics Reference a-Factor (F) XIIa Prekallikrein 1.8 lM Km; kcat/Km = 0.57 lM [87] FXI – [88] Complement C1 – [89] FVII – [90] Plasminogen – [91] High-Mr kininogen – [89] b-FXIIa Prekallikrein 2.1 lM Km; kcat/Km = 1.67 lM [87] Autoactivation of FXII FXII 2.4 lM Km; kcat/Km = 0.02 lM [92] Plasma kallikrein FXII 11 lM Km; kcat/Km = 0.57 lM [88] Single-chain urokinase 0.064 lM Km [22] High-Mr kininogen 1.4 lM Km; kcat/Km = 0.46 lM [93] Prolylcarboxypeptidase Prekallikrein 0.007 lM Km [35] Table 2 Inhibitors of the enzymes of the plasma kallikrein–kinin system independently of HK [22–24]. FXI also binds to prothrombin Enzyme Inhibitor Inhibition constant+ Reference and the glycoprotein Iba–IX–V complex on platelets [24,25]. Membrane-binding proteins of HK include gC1qR, urokinase a-Factor C1 inhibitor 222.0 · 103 M)1 min)1 [9] plasminogen activator receptor (u-PAR), and cytokeratin 1 (F) XIIa a2-Antiplasmin 11.0 · 103 M)1 min)1 [9] (CK1) (Fig. 1) [26–29]. When HK is proteolyzed by plasma a2-Macroglobulin 5.3 · 103 M)1 min)1 [9] Antithrombin 1.3 · 103 M)1 min)1 [9] kallikrein or other proteases to form cleaved HK (HKa), Plasma C1 inhibitor 102.0 · 104 M)1 min)1 [9] membrane tropomysin also functions as a binding site uniquely kallikrein a2-Macroglobulin 69.0 · 104 M)1 min)1 [10] for this form of kininogen [30]. FXII also has been shown to Antithrombin 1.8 · 104 M)1 min)1 [10] bind to gC1qR, u-PAR, and CK1 [27,31]. Both PK and FXI a1-Antitrypsin 0.025 · 104 M)1 min)1 [10] circulate in plasma almost completely bound to HK, but PK PAI-1 360.0 · 104 M)1 min)1 [94] Protein C inhibitor 600.0 · 104 M)1 min)1 [95] binding to endothelial cells predominates [24]. The reasons for this are as follows: (i) the concentration of PK (450 nM) is more PAI-1, plasminogen activator inhibitor-1. + than tenfold greater than that of FXI (30 nM) in plasma; and The values are second-order rate constants. (ii) the free Zn2+ concentration required for PK binding is only 0.3 lM, whereas that for FXI binding is 7 lM [24]. ordering of the adsorbed protein molecules that lead to When HK and PK assemble on endothelial cells and matrix, expression of its active site [13]. Negatively charged surfaces plasma kallikrein activity arises independently of added FXIIa. consist of artificial materials as found in coagulation assays such This event occurs in the presence of neutralizing antibody to as kaolin, celite, and glass surfaces. Several physiologic substances, such as articular cartilage, skin, fatty acids, endo- toxin, sodium urate crystals, calcium pyrophosphate, L-homo- cysteine, hematin, protoporphyrin, sulfatides, heparins, chondrotin sulfates, and amyloid b-protein, also support autoactivation of FXII. Formation of activated FXII by autoactivation results in PK activation with reciprocal activa- tion of FXII and PK and activation amplification of the system. In vivo, FXII autoactivation occurs on developing thrombus, contributing to its extent [7]. Substances that contribute to Ôcontact activationÕ on a developing thrombus include RNA from degrading cells, polysomes from platelet membranes, and fibrin itself [14,15]. FXII activation also occurs under conditions of sepsis, where bacteria provide a negatively charged surface, proteases to activate FXII, or a binding site [16,17]. Fig. 1. Physiologic assembly and activation of the plasma kallikrein–kinin system. The high molecular weight kininogen (HK)–prekallikrein (PK) Constitutive activation of the plasma KKS in the complex binds to its HUVEC receptor complex, which includes cytoker- atin 1 (CK1), urokinase plasminogen activator receptor (u-PAR) and intravascular compartment gC1qR. Prolylcarboxypeptidase (PRCP) bound to the complex activates It has been recognized that HK, FXII and PK specifically, PK to form plasma kallikrein (KAL). The KAL cleaves HK and acti- vates FXII and single-chain urokinase plasminogen activator (Scu-PA). saturably and reversibly bind to endothelial cells, platelets Cleaved HK liberates bradykinin (BK), which is a potent activator of and granulocytes [18–21]. HK serves as the major binding site tissue-type plasminogen activator (t-PA), NO (nitric oxide) and prosta- for PK and FXI, although both bind to endothelial cells cyclin (PGI2) liberation from endothelial cells. HKa, cleaved HK. Ó 2007 International Society on Thrombosis and Haemostasis
  • 3. The changing kallikrein–kinin system 2325 FXII and FXII-deficient plasma, but not PK-deficient plasma Vascular activities of the plasma KKS [22,32]. The plasma kallikrein formed results in kinetically favorable single-chain urokinase activation (Km = 64 nM) Regulation of blood pressure and flow Local BK formation (Table 1) [22]. The plasma kallikrein on endothelial cells also is known to influence blood pressure. BK is liberated from HK results in kinetically favorable FXII activation [33]. These data by plasma or tissue kallikrein cleavage. The nine amino acid provide an alternative hypothesis to contact activation for BK peptide, RPPGFSPFR, has two intravascular receptors: FXIIa formation in vivo. The increased requirements for free B2R, which is constitutively expressed, and the BK B1 receptor Zn2+ for FXII binding to endothelial cells suggest that FXIIÕs (B1R), which becomes expressed in inflammatory states. BK association and activation on endothelial cells follows HK and binds to B2R, a seven-transmembrane G-protein-coupled PK assembly and activation [24,31]. This proposed mechanism receptor, and stimulates its G-proteins to release nitric oxide for PK activation in vivo may be occurring constitutively. (NO), prostaglandin I2 (prostacyclin), smooth muscle Firstly, C1INH knockout mice have constitutive tissue edema hyperpolarization factor, and superoxide [43–46]. In sepsis, due to increased BK, as it is blocked by a B2R antagonist or by excessive BK release contributes to hypotension. mating C1INH and B2R knockout mice [11]. As plasma BK BK produced by the plasma and tissue KKS influences only arises from plasma kallikrein formation and C1INH only cardiovascular physiology. B2R knockout mice are not consti- inhibits plasma kallikrein, not tissue kallikein, BK must be tutively hypertensive; however, upon being subjected to a salt constantly formed in vivo to give the paw edema seen [11]. load, they have early-onset salt-sensitive hypertension [47]. B2R Secondly, FXII knockout mice also have plasma BK formation is involved in the control of regional vascular tone in the coro- without the presence of FXII [34]. nary arteries and the kidneys. The cardioprotective effects of A PK activator was purified from endothelial cells [35]. On angiotensin-converting enzyme (ACE) inhibition, which inhibits amino acid sequencing, it was identified as the serine protease BK degradation, is lost in B2R knockout mice. In diabetic mice, prolylcarboxypeptidase (PRCP) [35]. The Km of PRCP activa- the absence of B2R increases oxidative stress, mitochondrial tion of plasma PK (Km = 7 nM) is two hundred and fifty- to DNA damage, and senescence-associated phenotypes [48]. In three hundredfold higher than that for activated forms of FXII tissue kallikrein knockout mice, with reduced tissue BK (Table 1). This suggests that PRCP activation of PK is favored formation, there is thinning of the septum and posterior wall over that of a-FXIIa or b-FXIIa in vivo (Table 1). It is of note of the heart, resulting in ventricular dilatation and reduced left that C1INH is a tighter inhibitor of plasma kallikrein than of ventricular mass [49]. Furthermore, genetic kininogen deficiency activated FXII, suggesting that plasma kallikrein regulation is in rats contributes to aortic aneurysm formation [50]. more important than that of FXIIa (Table 2). PRCP was first recognized as a degrading enzyme for BK and angiotensin II Thrombosis risk Emerging information indicates that the (Ki 1 and 0.15 mM, respectively) by cleaving Pro-X bonds on plasma KKS influences thrombosis risk independently of the C-terminus of the protein [36]. Both purified and hemostasis [7,8]. Patients with FXII, PK and HK deficiency are recombinant PRCP activate PK with a Km 7–17 nM exceedingly rare, and although they do not bleed, there are too [35,37]. Although thought to be lysosomal in origin, PRCP is few patients to characterize a common clinical phenotype. a membrane and matrix protein, as it can be demonstrated to be FXII deficiency is more common than HK or PK deficiency. there functionally and immunochemically and it was interrupted Clinical investigations for venous thrombosis risk or on by a gene trap targeted to membrane proteins [35,37–39]. PRCP polymorphisms of FXII and their influence on cardiovascular is a risk factor for metabolic syndrome in men, and a PRCP disease have been conflicting (see below). The clearest polymorphism is associated with pre-eclampsia in women information on thrombosis risk or risk amelioration has been [40,41]. CHO cells with overexpressed PRCP have increased derived from animal models, which demonstrate unexpected PK-activating activity over controls; treatment of these cells findings. with small interfering RNA reduces the PK activation on these cells [42]. Finally, transfected CHO cells mostly express PRCP BK and kininogen BK infusion is a potent stimulant for tissue- on their membranes. These combined studies indicate that there type plasminogen activator (t-PA) release in rabbits and humans is a constitutive, physiologic endothelial cell mechanism for PK [51]. Kininogen itself has been shown to have antithrombin activation independent of FXII autoactivation by contact. activities. Both HK and low molecular weight kininogen at 5% of their physiologic concentrations block thrombin-induced platelet aggregation and serotonin release by inhibiting Activities of the plasma KKS thrombin binding to platelets [52]. The thrombin inhibitory The studies described above reveal a means for KKS assembly regions of kininogen have been associated with domains 3 and 4, and activation by physiologic and pathophysiologic mecha- the BK region [53,54]. A peptide comprising the first five amino nisms. Several vascular and cellular activities derive from these acids of BK, RPPGF, was found to bind weakly to the active site pathways. KKS vascular activities include regulation of blood of thrombin upon cocrystallization, and to bind the exodomains pressure and flow and thrombosis risk; the cellular activities of protease-activated receptor (PAR)1 and PAR4 to prevent include cellular proliferation, growth, angiogenesis, apoptosis, thrombin cleavage [55,56]. RPPGF inhibits in vitro and, when and inflammation. infused in dogs and humans, ex vivo thrombin-induced platelet Ó 2007 International Society on Thrombosis and Haemostasis
  • 4. 2326 A. H. Schmaier and K. R. McCrae aggregation [57,58]. RPPGF in pharmacologic doses prevents binds to the overexpressed angiotensin receptor 2 to increase carotid artery thrombosis in mice and coronary artery NO and prostacyclin, and prolong the bleeding time of the thrombosis in dogs [57,59,60]. animal [8] (Fig. 2). Thirdly, RPPGF is elevated in these animals, As BK induces NO, prostacyclin and t-PA release from due to increased BK degradation by ACE [8]. The elevation of endothelial cells, we hypothesized that the B2R knockout mouse RPPGF levels may also contribute to the thrombosis protection. would be prothrombotic. To our surprise, B2R knockout mice These combined studies indicate that BK and its receptor system have delayed carotid artery occlusion times in the Rose Bengal indirectly influence thrombosis risk by influencing endothelial model (Fig. 2) [8]. The mechanism for thrombosis protection is cell biology through cross-talk with components of the plasma dependent on this systemÕs interaction with the renin–angioten- RAS. Such a pathway for risk modification of intra-arterial sin system (RAS) [61]. In the RAS, angiotensinogen is converted thrombosis has not been previously appreciated. to angiotensin I by renin and then converted to angiotensin II by ACE. ACE also is the major enzyme that degrades BK to BK FXII There are conflicts between human clinical and 1–5 (RPPGF) in the intravascular compartment (Fig. 2). experimental animal data for the role of FXII in thrombosis Angiotensin II usually binds to angiotensin receptor 1 to induce risk. A polymorphism in FXII (46C/T) is associated with vasoconstriction and salt retention, and elevate blood pressure. increased risk for arterial thrombosis [63–65]. Individuals However, if angiotensin receptor 2 is overexpressed, angiotensin homozygous for the 46C/T polymorphism have lowered FXII II will preferentially bind to it to induce vasodilatation and and FXIIa levels. Reduced activated forms of FXII may be blood pressure reduction. The mechanism by which the B2R associated with reduced total fibrinolytic activity, resulting in knockout mice are protected from thrombosis is 3-fold. Firstly, increased thrombosis risk. This interpretation is opposite to in the absence of B2R, angiotensin receptor 2 is overexpressed what is demonstrated in FXII-deficient mice [7]. FXII-deficient (Fig. 2). B2R and angiotensin receptor 2 colocalize in cells, and mice have reduced thrombus after induction of arterial clots there is an as yet unrecognized mechanism whereby the presence [7,66]. The mechanism for the increased size of thrombus in of one GPCR receptor regulates the expression of the other mice that have normal levels of FXII may be related to [8,62]. Secondly, there is increased angiotensin II as a result of increased contact activation occurring on a developing platelet reduced BK uptake into cells with reflexive increased ACE thrombus [14,15]. Therapeutic inhibition of FXII may result in degradative activity [8] (Fig. 2). The increased angiotensin II reduced thrombus formation without bleeding. These observations were not predicted by in vitro investigations on the biochemistry and cell biology of FXII and clinical studies on populations with polymorphisms or defects in FXII. Cellular activities of the plasma KKS Cell proliferation and angiogenesis Investigations have shown that kininogen and related proteins influence cellular activities of endothelial and other cells. These investigations were prompted by the observation that HKa induces selective apoptosis of proliferating endothelial cells and inhibits angiogenesis [67,68]. HKa inhibits neovascularization of s.c. planted Matrigel plugs, as well as fibroblast growth factor 2-induced angiogenesis in the chick chorioallantoic membrane Fig. 2. Mechanisms for thrombosis protection in bradykinin B2 receptor assay [67,68]. Moreover, peptides from domain 5 of HK (D5), (B2R) knockout mice. In the absence of B2R, there is increased plasma which subsumes the HK cell-binding region, induce endothelial bradykinin, as B2R accounts for 40% of the metabolism of bradykinin. cell apoptosis, inhibit angiogenesis, and are antibacterial Increased bradykinin results in increased conversion to bradykinin 1–5 (peptide RPPGF) (Blood 2006; 108: 192–99). As a byproduct of increased [69–71]. Kininogen-deficient Brown Norway Katholiek rats, RPPGF formation, there are increased levels of angiotensin II (Blood alternatively, display decreased angiogenesis, possibly resulting 2006; 108: 192–99). Angiotensin-converting enzyme (ACE) also converts from deficient BK release that is ameliorated by a BK analog or angiotensin I to angiotensin II. In the absence of B2R, there is increased kininogen replacement [72,73]. The mechanism(s) by which expression of the angiotensin receptor 2 (AT2R). The increased angio- these activities occur is not known, but may involve the anti- tensin II is shunted to overexpressed AT2R, as angiotensin II has the same binding affinity for angiotensin receptor 1 and AT2R. This leads to a adhesive function of HKa towards cells on vitronectin, the paradoxical effect in comparison to the usual angiotensin II elevation. kininogen multiprotein receptor complex, or tropomyosin Increased stimulation of AT2R produces vasodilatation and increased [30,74,75]. plasma nitric oxide (NO) and prostacyclin (PGI2) (Blood 2006; 108: 192– 99). The increased NO and PGI2 prolong the bleeding time, and these Outside-in signaling mediated by the KKS Although the animals have delayed thrombosis risk on the Rose Bengal model for carotid artery thrombosis. These investigations indicate that thrombosis proangiogenic activities of the KKS are mediated by B1R and risk can be modified by factors independent of coagulation, fibrinolytic or B2R, a different receptor system(s) may be involved in the anticoagulant proteins. inhibition of cell proliferation, adhesion, anti-apoptosis and Ó 2007 International Society on Thrombosis and Haemostasis
  • 5. The changing kallikrein–kinin system 2327 angiogenesis. u-PAR has been shown to mediate intracellular 4 Ratnoff OD, Davie EW. Waterfall sequence for intrinsic blood clot- signaling. u-PA binding to u-PAR triggers a cascade of ting. Science 1964; 145: 1310–2. 5 Weuppers KD, Cochrane CG. Plasma prekallikrein:isolation, charac- intracellular tyrosine phosphorylation that includes src-type terization, and mechanism of action. J Exp Med 1972; 135: 1–20. protein kinases, focal adhesion proteins, p38, and extracellular 6 Colman RW, Bagdasarian A, Talamos RC, Seavey M, Scott CF, signal-related kinase 1/2 (ERK1/2) [76–78]. When HKa or D5 Kaplan AP. Williams trait. Human kininogen deficiency with dimin- peptides bind to cultured endothelial cells after basic fibroblast ished levels of plasminogen proactivator and prekallikrein associated growth factor-induced cell proliferation, there is a reduction of with abnormalities of the Hageman factor-dependent pathways. J Clin Invest 1975; 56: 1650–62. cyclin D1 expression and an upregulation of Cdc2 and cyclin A 7 Renne T, Pozgajova M, Gruner S, Schuh K, Pauer HU, Burfeind P, [79,80]. HKa also inhibits adhesion of endothelial cells to Gailani D, Nieswandt B. Defective thrombus formation in mice vitronectin (90%) and gelatin (40%) without any effect on lacking coagulation factor XII. J Exp Med 2005; 280: 28572–80. adhesion to fibronectin, on which it induces endothelial cell 8 Shariat-Madar Z, Mahdi F, Warnock M, Homeister JW, Srikanth S, apoptosis. Endothelial cell migration induced by sphingosine Krijanovski Y, Murphey LJ, Jaffa AA, Schmaier AH. Bradykinin B2 receptor knockout mice are protected from thrombosis by increased 1-phosphate and vascular endothelial growth factor is blocked nitric oxide and prostacyclin. Blood 2006; 108: 192–9. by HKa and D5 and may be associated with inhibition of 9 Pixley RA, Schapira M, Colman RW. The regulation of human phosphorylation of phosphinositide 3 (PI3)-kinase-Akt and factor XIIa by plasma proteinase inhibitors. J Biol Chem 1985; 260: glycogen synthase kinase (GSK)-3a [81]. 1723–9. u-PAR is known to upregulate Mac-1 adhesion to fibrinogen, 10 Schapira M, Scott CF, Colman RW. Protection of human plasma kallikrein from inactivation by C1 inhibitor and other proteases. The and focal adhesion kinase (FAK) and mitogen-activated protein role of high molecular weight kininogen. Biochemistry 1981; 20: 2738– kinase are involved in this process [82]. Furthermore, u-PAR 43. binds vitronectin and HKa, and D5 disrupts u-PAR–integrin 11 Han ED, MacFarlane RC, Mulligan AN, Scafidi J, Davis AE III. and u-PAR–vitronectin interactions [74,83]. FXII activates Increased vascular permeability in C1 inhibitor-deficient mice medi- ERK1/2 in HEPG2 cells and cultured vascular smooth muscle ated by the bradykinin type 2 receptor. J Clin Invest 2002; 109: 1057– 63. cells [84]. Vitronectin binds to the same region on u-PAR as HK, 12 Wiggins RC, Cochran CG. The autoactivation of rabbit Hageman HKa, single-chain urokinase plasminogen activator (Scu-PA), factor. J Exp Med 1979; 150: 1122–33. and FXII (unpublished) [75,85,86]. Preliminary studies indicate 13 Chen X, Wang J, Paszti Z, Wang F, Schrauben JN, Tarabara VV, that Scu-PA or FXII upregulate ERK1/2 and Akt on cultured Schmaier AH, Chen Z. Ordered adsorption of coagulation factor XII endothelial cells, whereas HKa and D5-derived peptides as well on negatively charged polymer surfaces probed by sum frequency gen- eration vibrational spectroscopy. Anal Bioanal Chem 2007; 388: 65–72. as peptides from domain 2 of u-PAR block this interaction [86]. 14 Kannemeier C, Shibamiya A, Nakazawa F, Trusheim H, Ruppert C, The upregulation of ERK1/2 and Akt is mediated by a Markart P, Song Y, Tzima E, Kennerknecht E, Niepmann M, von b1-integrin, is independent of lipid rafts, and is associated with Bruehl ML, Sedding D, Massberg S, Gunther A, Engelmann B, increased endothelial cell proliferation and incorporation of Preissner KT. Extracellular RNA constitutes a natural procoagulant 5-bromo-2¢-deoxy-uridine [86]. A u-PAR-mediated signaling cofactor in blood coagulation. Proc Natl Acad Sci USA 2007; 104: 6388–93. system may be the additional pathway leading to cell prolifer- 15 Smith SA, Mutch NJ, Baskar D, Rohloff P, Docampo R, Morrissey ation, growth and, perhaps, apoptosis and angiogenesis JH. Polyphosphate modulates blood coagulation and fibrinolysis. Proc modulated by HK and its cleavage products. Natl Acad Sci USA 2006; 103: 903–8. 16 Matsumoto K, Yamamoto T, Kamata R, Maeda H. Pathogenesis of serratial infection: activation of the Hageman–prekallikrein cascade by Acknowledgements serratial protease. J Biochem 1984; 96: 739–49. 17 Herwald H, Morgelin M, Olsen A, Rhen M, Dahlback B, Muller- This work was supported in part by grants HL052779, Esterl W. Activation of the contact-phase system on bacterial surfaces HL055709 and HL086038 to A. H. Schmaier, and grants is a clue to serious complications in factious disease. Nat Med 1998; 4: HL076810, CA83134 and P50HL081011 to K. R. McCrae. 298–302. 18 Gustafson EG, Schutsky D, Knight L, Schmaier AH. High molecular weight kininogen binds to unstimulated platelets. J Clin Invest 1986; Disclosure of Conflict of Interests 78: 310–8. 19 Gustafson EJ, Schmaier AH, Wachtfogel YT, Kaufman N, Kucich U, The authors state that they have no conflict of interest. Colman RW. Human neutrophils contain and bind high molecular weight kininogen. J Clin Invest 1989; 84: 28–25. 20 Schmaier AH, Kuo A, Lundberg D, Murray SC, Cines DB. Expres- References sion of high molecular weight kininogen on human umbilical vein endothelial cells. J Biol Chem 1988; 263: 16327–33. 1 Sainz IM, Pixley RA, Colman RA. Fifty years of research on the 21 Reddigari SR, Shibayama Y, Brunnee T, Kaplan AP. Human Hag- plasma kallikrein–kinin system: from protein structure and function to eman factor (factor XII) and high molecular weight kininogen compete cell biology and in-vivo pathophysiology. Thromb Haemost 2007; 98: for the same binding site on human umbilical vein endothelial cells. 77–83. J Biol Chem 1993; 268: 11982–7. 2 Gailani D, Renne T. The intrinsic pathway of coagulation: a target for 22 Motta G, Rojkjaer R, Hasan AAK, Cines DB, Schmaier AH. High treating thromboembolic disease. J Thromb Haemost 2007; 5: 1106–12. molecular weight kininogen regulates prekallikrein assembly and 3 Ratnoff OD, Colopy JE. A familial hemorrhagic trait associated with a activation on endothelial cells: a novel mechanism for contact activa- deficiency of a clot promoting fraction of plasma. J Clin Invest 1955; tion. Blood 1998; 91: 516–28. 34: 602–13. Ó 2007 International Society on Thrombosis and Haemostasis
  • 6. 2328 A. H. Schmaier and K. R. McCrae 23 Shariat-Madar Z, Mahdi F, Schmaier AH. Factor XI assembly and 42 Shariat-Madar Z, Rahimi E, Mahdi F, Schmaier AH. Over-expression activation on human umbilical vein endothelial cells in culture. Thromb of prolylcarboxypeptidase enhances plasma prekallikrein activation on Haemost 2001; 85: 544–51. Chinese hamster ovary cells. Am J Physiol Heart Circ Physiol 2005; 24 Mahdi F, Shariat-Madar Z, Schmaier AH. The relative priority of 289: H2697–703. prekallikrein and factors XI/XIa assembly on cultured endothelial 43 Palmer RMJ, Ferrige AG, Moncada S. Nitric oxide release accounts cells. J Biol Chem 2003; 278: 43983–90. for the biologic activity of endothelium-derived relaxing factor. Nature 25 Yun TH, Baglia FA, Myles T, Navaneetham D, Lopez JA, Walsh PN, 1987; 372: 524–6. Leung LL. Thrombin activation of factor XI on activated platelets 44 Hong SL. Effect of bradykinin and thrombin on prostacyclin synthesis requires the interaction of factor XI and platelet glycoprotein Ib alpha in endothelial cells from calf and pig aorta and human umbilical cord with thrombin anion-binding exosites I and II, respectively. J Biol vein. Thromb Res 1980; 18: 787–95. Chem 2003; 278: 48112–9. 45 Feletou M, Vanhoutte PM. Endothelium-derived hyperpolarization 26 Herwald H, Dedio J, Kellner R, Loos M, Muller-Esterl W. Isolation factor: where are we now? Arterioscler Thromb Vasc Biol 2006; 26: and characterization of the kininogen binding protein p33 from 1215–25. endothelial cells. J Biol Chem 1996; 271: 13040–7. 46 Holland JA, Pritchard KA, Pappolla MA. Bradykinin induces 27 Joseph K, Ghebrehiwet B, Peerschke EIB, Reid KBM, Kaplan AP. superoxide anion release from human endothelial cells. J Cell Physiol Identification of the zinc-dependent endothelial cell binding protein for 1990; 143: 21–5. high molecular weight kininogen and factor XII: identity with the 47 Cervenka L, Harrison-Bernard LM, Dipp S, Primrose G, Imig JD, El- receptor that binds to the globular ÔheadsÕ of C1q (qC1qR). Proc Natl Dahr SS. Early onset salt-sensitive hypertension in bradykinin B2 Acad Sci USA 1996; 93: 8552–7. receptor null mice. Hypertension 1999; 34: 176–80. 28 Colman RW, Pixley RA, Najamunnisa S, Yan W, Wang J, Mazar A, 48 Kakoki M, Kizer CM, Yi X, Takahashi N, Kim H-S, Bagnell CR, McCrae KR. Binding of high molecular weight kininogen to human Edgell C-J, Maeda N, Jennette JC, Smithies O. Senescence-associated endothelial cells is mediated via a site within domains 2 and 3 of the phenotypes in Akita diabetic mice are enhanced by absence of urokinase receptor. J Clin Invest 1997; 100: 1481–7. bradykinin B2 receptors. J Clin Invest 2006; 116: 1302–9. 29 Hasan AAK, Zisman T, Schmaier AH. Identification of cytokeratin 1 49 Meneton P, Bloch-Faure M, Hagege AA, Ruetten H, Huang W, as a binding protein and presentation receptor for kininogens on Bergaya S, Ceiler D, Gehring D, Martins I, Salmon G, Boulanger C, endothelial cells. Proc Natl Acad Sci USA 1998; 95: 3615–20. Nussberger J, Crozatier B, Gasc J-M, Heudes D, Bruneval P, 30 Zhang J-C, Donate F, Qi X, Ziats NP, Juarez JC, Mazar AP, Pang Y- Doetschman T, Menard J, Alhenc-Gelas F. Cardiovascular P, McCrae KR. The antiangiogenic activity of cleaved high molecular abnormalities with normal blood pressure in tissue kallikrein-deficient weight kininogen is mediated through binding to endothelial cell mice. Proc Natl Acad Sci USA 2001; 98: 2634–9. tropomyosin. Proc Natl Acad Sci USA 2002; 99: 12224–9. 50 Kaschina E, Stoll M, Sommerfeld M, Steckelings UM, Kreutz R, 31 Mahdi F, Shariat-Madar Z, Figueroa CD, Schmaier AH. Factor XII Unger T. Genetic kininogen deficiency contributes to aortic aneu- interacts with the multiprotein assembly of urokinase plasminogen rysm formation but not to atherosclerosis. Physiol Genomics 2004; 19: activator receptor, gC1qR, and cytokeratin on endothelial cell mem- 41–9. branes. Blood 2002; 99: 3585–96. 51 Brown NJ, Gainer JV, Stein CM, Vaughan DE. Bradykinin stimulates 32 Motta G, Shariat-Madar Z, Mahdi F, Sampaio CAM, Schmaier AH. tissue plasminogen activator release in human vasculature. Hyperten- Assembly and activation of high molecular weight kininogen and sion 1999; 33: 1431–5. prekallikrein on cell matrix. Thromb Haemost 2001; 86: 840–7. 52 Meloni FJ, Schmaier AH. Low molecular weight kininogen binds to 33 Rojkjaer R, Hasan AAK, Motta G, Schousboe I, Schmaier AH. platelets to modulate thrombin-induced platelet activation. J Biol Factor XII does not initiate prekallikrein activation on endothelial Chem 1991; 266: 6786–94. cells. Thromb Haemost 1998; 80: 74–81. 53 Jiang Y, Muller-Esterl W, Schmaier AH. Domain 3 of kininogens 34 Iwaki T, Castellino FJ. Plasma levels of bradykinin are suppressed in contains a cell binding site and a site that modifies thrombin activation factor XII-deficient mice. Thromb Haemost 2006; 95: 1003–10. of platelets. J Biol Chem 1992; 267: 3712–7. 35 Shariat-Madar Z, Mahdi F, Schmaier AH. Identification and char- 54 Hasan AAK, Amenta S, Schmaier AH. Bradykinin and its metabolite acterization of prolylcarboxypeptidase as an endothelial cell prekal- ARG-PRO-PRO-GLY-PHE are selective inhibitors of a-thrombin- likrein activator. J Biol Chem 2002; 277: 17962–9. induced platelet activation. Circulation 1996; 94: 517–28. 36 Oyda CE, Marinkovic DV, Hammon KJ, Stewart TA, Erdos EG. 55 Hasan AAK, Warnock M, Nieman M, Srikanth S, Mahdi F, Krish- Purification and properties of prolylcarboxypeptidase (angiotensinase nan R, Tulinsky A, Schmaier AH. The mechanisms of Arg-Pro-Pro- C) from human kidney. J Biol Chem 1978; 253: 5927–31. Gly-Phe inhibition of thrombin. Amer J Physiol Heart and Circ Physiol 37 Shariat-Madar Z, Mahdi F, Schmaier AH. Recombinant prolyl- 2003; 285: H183–93. carboxypeptidase activates plasma prekallikrein. Blood 2004; 103: 56 Nieman MT, Pagan-Ramos E, Warnock M, Krijanovski Y, Hasan 4554–61. AAK, Schmaier AH. Mapping the interaction of bradykinin 1–5 with 38 Moreira CR, Schmaier AH, Mahdi F, da Motta G, Nader HB, Sha- the exodomain of protease activated receptor 4 (PAR4). FEBS Lett riat-Madar Z. Identification of prolylcarboxypeptidase as the cell 2005; 579: 25–9. matrix-associated prekallikrein activator. FEBS Lett 2002; 523: 167– 57 Hasan AAK, Rebello SS, Smith E, Srikanth S, Werns S, Driscoll E, 70. Faul J, Brenner D, Normolle D, Lucchesi BR, Schmaier AH. 39 Skarnes WC. Gene trapping methods for the identification and func- Thrombostatin inhibits induced canine coronary thrombosis. Thromb tional analysis of cell surface proteins in mice. Methods Enzymol 2000; Haemost 1999; 82: 1182–7. 328: 592–615. 58 Murphey LJ, Malave HA, Petro J, Biaggioni I, Byrne DW, Vaughan 40 McCarthy JJ, Meyer J, Moliterno DJ, Newby LK, Rogers WJ, Topol DE, Luther JM, Pretorius M, Brown NJ. Bradykinin and its metab- EJ, GenQuest multicenter study. Evidence for substantial effect mod- olite bradykinin 1–5 inhibit thrombin-induced platelet aggregation in ification by gender in a large scale genetic association study of the humans. J Pharm Exp Ther 2006; 318: 1287–92. metabolic syndrome among coronary heart disease patients. Hum 59 Nieman MT, Warnock M, Hasan AAK, Mahdi F, Lucchesi BR, Genet 2003; 114: 87–98. Brown NJ, Murphey LJ, Schmaier AH. The preparation and char- 41 Wang L, Feng Y, Zhang Y, Zhou H, Jiang S, Niu T, Wei LJ, Xu X, Xu acterization of novel peptide antagonists to thrombin, factor VIIa and X, Wang X. Prolylcarboxypeptidase gene, chronic hypertension, and activation of protease activates receptor 1. J Pharm Exp Ther 2004; risk of preeclampsia. Am J Obstet Gynecol 2006; 195: 162–71. 311: 492–501. Ó 2007 International Society on Thrombosis and Haemostasis
  • 7. The changing kallikrein–kinin system 2329 60 Hasan AAK, Schmaier AH, Warnock M, Normolle D, Driscoll E, tion of focal adhesion proteins and activation of mitogen-activated Lucchesi BR, Werns SW. Thrombostatin inhibits cyclic flow variations protein kinase in cultured endothelial cells. J Biol Chem 1998; 273: in stenosed canine coronary arteries. Thromb Haemost 2001; 86: 1296– 18268–72. 304. 78 Nguyen DHD, Webb DJ, Catling AD, Song Q, Dhakephalkar A, 61 Schmaier AH. The kallikrein–kinin and the rennin–angiotensin sys- Weber MJ, Ravichandran KS, Gonias SL. Urokinase-type plasmin- tems have a multilayered interaction. Am J Physiol Regul Integr Comp ogen activator stimulates the Ras/Extracellular signal-regulated kinase Physiol 2003; 285: R1–13. (ERK) signaling pathway and MCF-7 cell migration by a mechanism 62 Abadir PM, Periasamy A, Carey RM, Siragy HM. Angiotensin II type that requires focal adhesion kinase, Src, and Shc. J Biol Chem 2000; 2 receptor–bradykinin B2 receptor functional heterodimerization. 275: 19382–8. Hypertension 2006; 48: 316–22. 79 Guo Y-L, Wang S, Colman RW. Kininostatin, an angiogenic inhibi- 63 Soria JM, Almasy L, Souto JC, Bacq D, Buil A, Faure A, Martinez- tor, inhibits proliferation and induces apoptosis of human endothelial Marchan E, Mateo J, Borrell M, Stone W, Lathrop M, Fontcuberta J, cells. Arterioscler Thromb Vasc Biol 2001; 21: 1427–33. Blangero J. A quantitative-trait locus in the human factor XII gene 80 Wang S, Hasham MG, Isordia-Salas I, Tsygankov AY, Colman RW, influences both plasma factor XII levels and susceptibility to throm- Guo Y-L. Upregulation of Cdc2 and cyclin A during apoptosis of botic disease. Am J Hum Genet 2002; 70: 567–74. endothelial cells induced by cleaved high-molecular-weight kininogen. 64 Zito F, Lowe GDO, Rumley A, McMahon AD, Humphries SE. Am J Physiol Heart Circ Physiol 2003; 284: H1917–23. Association of the factor XII 46CT polymorphism with risk of 81 Katkade V, Soyombo AA, Isordia-Salas I, Bradford HN, Gaughan coronary heart disease in the WOSCOPS study. Atherosclerosis 2002; JP, Colman RW, Panetti TS. Domain 5 of cleaved high molecular 165: 153–8. weight kininogen inhibits endothelial cell migration through Akt. 65 Colhoun HM, Zito F, Chan NN, Rubens MB, Fuller JH, Humphries Thromb Haemost 2005; 94: 606–14. SE. Activated factor XII levels and factor XII 46CT genotype in 82 Zhang H, Colman RW, Sheng N. Regulation of CD11b/CD18 (Mac- relation to coronary artery calcification in patients with type 1 diabetes 1) adhesion to fibrinogen by urokinase receptor (uPAR). Inflamm Res and healthy subjects. Atherosclerosis 2002; 163: 363–9. 2003; 52: 86–93. 66 Kleinschnitz C, Stoll G, Bendszuz M, Schuh K, Pauer HU, Burfeind P, 83 Cunningham O, Andolfo A, Santovito ML, Iuzzolino L, Blasi F, Renne C, Gailani D, Nieswandt B, Renne T. Targeting coagulation Sidenius N. Dimerization controls the lipid raft partitioning of uPAR/ factor XII provides protection from pathologic thrombosis in cerebral CD87 and regulates its biologic functions. EMBO J 2003; 22: 5994– ischemia without interfering with hemostasis. J Exp Med 2006; 203: 6003. 513–8. 84 Gordon EM, Venkatesan N, Salazat R, Tang H, Schmeidler-Sapiro K, 67 Zhang J-C, Claffey K, Sakthivel R, Darzynkiewicz Z, Shaw DE, Leal Buckley S, Warburton D, Hall FL. Factor XII-induced mitogenesis is J, Wang Y-C, Lu F-M, McCrae KR. Two-chain high molecular weight mediated via a distinct signal transduction pathway that activates a kininogen induces endothelial cell apoptosis and inhibits angiogenesis: mitogen-activated protein kinase. Proc Natl Acad Sci USA 1996; 93: partial activity within domain 5. FASEB J 2000; 14: 2589–600. 2174–9. 68 Colman RW, Jameson BA, Lin Y, Johnson D, Mousa SA. Domain 5 85 Li Y, Lawrence DA, Zhang L. Sequences within domain II of the of high molecular weight kininogen (kininostatin) down-regulates urokinase receptor critical for differential ligand recognition. J Biol endothelial cell proliferation and migration and inhibits angiogenesis. Chem 2003; 278: 29925–32. Blood 2000; 95: 543–50. 86 Schmaier AH, Mahdi F, Sitrin R. The urokinase plasminogen acti- 69 Zhang J-C, Qi X, Juarez J, Plunkett M, Donate F, Sakthivel R, Mazar vator receptor mediates ScuPA- or FXII-induced cell growth and AP, McCrae KR. Inhibition of angiogenesis by two-chain high proliferation. Blood 2006; 108(Suppl.): 1817 (abstract). molecular weight kininogen (HKa) and kininogen-derived polypep- 87 Tankersley DL, Finlayson JS. Kinetics of activation and autoactiva- tides. Can J Physiol Pharmacol 2002; 80: 85–90. tion of human factor XII. Biochemistry 1984; 23: 273–9. 70 Hasan AAK, Cines DB, Herwald H, Schmaier AH, Muller-Esterl W. 88 Kurachi K, Fujikawa K, Davie EW. Mechanism of activation of bo- Mapping the cell binding site on high molecular weight kininogenÕs vine factor XI by factor XII and factor XIIa. Biochemistry 1980; 19: domain 5. J Biol Chem 1995; 270: 19256–61. 1330–8. 71 Nordahl EA, Rydengard V, Morgelin M, Schmidtchen A. Domain 5 89 Ghrbrehiwet B, Randazzo BP, Dunn JT, Silverberg M, Kaplan AP. of high molecular weight kininogen is antibacterial. J Biol Chem 2005; Mechanisms of activation of the classical pathway of complement by 280: 34832–9. Hageman factor fragment. J Clin Invest 1983; 71: 1450–6. 72 Hu DE, Fan TP. [Leu8]des-Arg9-bradykinin inhibits the angiogenic 90 Seligsohn U, Osterud B, Brown SF, Rappaport SI. Activation of hu- effect of bradykinin and interleukin-1 in rats. Br J Pharmacol 1993; man factor VII in human plasma and purified systems. J Clin Invest 109: 14–7. 1979; 64: 239–43. 73 Hayashi I, Amano H, Yoshida S, Kamata K, Kamata M, Inukai M, 91 Goldsmith GH, Saito H, Ratnoff OD. The activation of plasminogen Fujita T, Kumagai Y, Furudat S, Majima M. Suppressed angiogenesis by Hageman factor (Factor XII) and Hageman Factor fragments. J in kininogen-deficiencies. Lab Invest 2002; 82: 871–80. Clin Invest 1978; 62: 54–60. 74 Chavakis T, Kanse SM, Lupu F, Hammes H-P, Muller-Esterl W, 92 Bernardo MM, Day DE, Olson ST, Shore JD. Surface-independent Pixley RA, Colman RW, Preissner KT. Different mechanisms define acceleration of factor XII activation by zinc ions. I Kinetic charac- the antiadhesive function of high molecular weight kininogen in inte- terization of the metal ion rate enhancement. J Biol Chem 1993; 268: grin- and urokinase receptor-dependent interactions. Blood 2000; 96: 12468–76. 514–22. 93 Tayeh MA, Olson ST, Shore JD. Surface-induced alterations in the 75 Mahdi F, Shariat-Madar Z, Kuo A, Carinato M, Cines DB, Schmaier kinetic pathway for cleavage of human high molecular weight kinin- AH. Mapping the interaction between high molecular weight kinino- ogen by plasma kallikrein. J Biol Chem 1994; 269: 16318–25. gen and the urokinase plasminogen activator receptor. J Biol Chem 94 Berrettini M, Schleef RR, Espana F, Loskutoff DJ, Griffin JH. 2004; 279: 16621–8. Interaction of type 1 plasminogen activator inhibitor with the 76 Konakova M, Hucho F, Schleuning W-D. Downstream targets of enzymes of the contact activation system. J Biol Chem 1989; 264: urokinase-type plasminogen-activator-mediated signal transduction. 11738–43. Eur J Biochem 1998; 253: 421–9. 95 Meijers JCM, Kanters DHA, Vlooswijk RRA, van Erp HE, Hessing 77 Tang H, Kerins DM, Hao Q, Inagami T, Vaughan DE. The urokinase- M, Bouma BN. Inactivation of human plasma kallikrein and factor type plasminogen activator receptor mediates tyrosine phosphoryla- Xia by protein C inhibitor. Biochemistry 1988; 27: 4231–7. Ó 2007 International Society on Thrombosis and Haemostasis