Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Thromb Haemost ; 105(6): 1053-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21544310

ABSTRACT

The physiologic activation of the plasma kallikrein-kinin system requires the assembly of its constituents on a cell membrane. High- molecular-weight kininogen (HK) and cleaved HK (HKa) both interact with at least three endothelial cell binding proteins: urokinase plasminogen activator receptor (uPAR), globular C1q receptor (gC1qR,) and cytokeratin 1 (CK1). The affinity of HK and HKa for endothelial cells are KD=7-52 nM. The contribution of each protein is unknown. We examined the direct binding of HK and HKa to the soluble extracellular form of uPAR (suPAR), gC1qR and CK1 using surface plasmon resonance. Each binding protein linked to a CM-5 chip and the association, dissociation and KD (equilibrium constant) were measured. The interaction of HK and HKa with each binding protein was zinc-dependent. The affinity for HK and HKa was gC1qR>CK1>suPAR, indicating that gC1qR is dominant for binding. The affinity for HKa compared to HK was the same for gC1qR, 2.6-fold tighter for CK1 but 53-fold tighter for suPAR. Complex between binding proteins was only observed between gC1qR and CK1 indicating that a binary CK1-gC1qR complex can form independently of kininogen. Although suPAR has the weakest affinity of the three binding proteins, it is the only one that distinguished between HK and HKa. This finding indicates that uPAR may be a key membrane binding protein for differential binding and signalling between the cleaved and uncleaved forms of kininogen. The role of CK1 and gC1qR may be to initially bind HK to the membrane surface before productive cleavage to HKa.


Subject(s)
Keratins/metabolism , Kininogen, High-Molecular-Weight/metabolism , Membrane Glycoproteins/metabolism , Receptors, Complement/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Surface Plasmon Resonance , Blood Coagulation , Endothelium/metabolism , Humans , Keratin-1/metabolism , Keratins/chemistry , Kininogen, High-Molecular-Weight/chemistry , Membrane Glycoproteins/chemistry , Protein Binding , Receptors, Complement/chemistry , Receptors, Urokinase Plasminogen Activator/chemistry , Signal Transduction
2.
J Thromb Haemost ; 5(2): 403-11, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17155949

ABSTRACT

BACKGROUND: Domain 5 (D5) of kininogen inhibits endothelial cell adhesion, migration, proliferation and angiogenesis by inducing apoptosis and disrupting a signaling pathway initiated by binding to the urokinase receptor (uPAR). OBJECTIVES: Because tumor cells frequently overexpress uPAR, we hypothesized that D5 can directly inhibit proliferation of colon carcinoma cells. METHODS AND RESULTS: A recombinant fusion protein of D5 and glutathione S-transferase (GST-D5) but not GST at 280 nm inhibited proliferation of human colon carcinoma cells (HCT-116) in vitro by 75-86%. We found that treatment with GST-D5 did not affect the survival pathway, phosphatidylinositol 3-kinase or the apoptotic pathway. In contrast, the G1/S phase transition of the cell cycle was downregulated as evidenced by an increase of cells in G0/G1 and a decrease in cells in S by flow cytometry. We found a decrease in serine phosphorylation of the retinoblastoma protein Rb (p107) after incubation with GST-D5. Less E2F-1 transcription factor and p107 were released and fewer cells overcame the G1/S growth restriction point. Expression levels of cyclins D1, A and E were reduced as measured by densiometric analysis of western blots. Cyclin-dependent protein kinase activities were downregulated and p27, the cyclin-dependent kinase inhibitor, was activated by GST-D5. CONCLUSIONS: These findings indicate that D5 of high molecular weight kininogen interferes with the G1 to S phase transition, reducing the proliferation of human colon carcinoma cells.


Subject(s)
Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Interphase/drug effects , Kininogens/physiology , Peptide Fragments/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/analysis , Cell Line, Tumor , Colonic Neoplasms/drug therapy , G1 Phase , Humans , Kininogens/chemistry , Recombinant Fusion Proteins/pharmacology , Retinoblastoma Protein/metabolism , S Phase
3.
FASEB J ; 18(7): 887-9, 2004 May.
Article in English | MEDLINE | ID: mdl-15001555

ABSTRACT

Bradykinin (BK), a vasoactive, proinflammatory nonapeptide, promotes cell adhesion molecule (CAM) expression, leukocyte sequestration, inter-endothelial gap formation, and protein extravasation in postcapillary venules. These effects are mediated by bradykinin-1 (B1R) and-2 (B2R) receptors. We delineated some of the mechanisms by which BK could influence chronic inflammation by altering CAM expression on leukocytes, endothelium, and synovium in joint sections of peptidoglycan-polysaccharide-injected Lewis rats. Blocking B1R results in significantly increased joint inflammation. Immunohistochemistry of the B1R antagonist group revealed increased leukocyte and synovial CD11b and CD54 expression and increased CD11b and CD44 endothelial expression. B2R antagonism decreased leukocyte and synovial CD44 and CD54 and endothelial CD11b expression. Although these findings implicate B2R involvement in the acute phase of inflammation by facilitating leukocyte activation (CD11b), homing (CD44), and transmigration (CD54). Treatment with a B2R antagonist did not affect the disease evolution in this model. In contrast, when both BK receptors are blocked, the aggravation of inflammation by B1R blockade is neutralized and there is no difference from the disease-untreated model. Our findings suggest that B1R and B2R signaling show physiologic antagonism. B1R signaling suggests involvement in down-regulation of leukocyte activation, transmigration, and homing. Further studies are needed to evaluate the B1 receptor agonist's role in this model.


Subject(s)
Arthritis/metabolism , Bradykinin/physiology , Cell Adhesion Molecules/biosynthesis , Endothelium, Vascular/drug effects , Leukocytes/drug effects , Synovial Membrane/drug effects , Animals , Arthritis/chemically induced , Arthritis/genetics , Arthritis, Rheumatoid/metabolism , Bradykinin/analogs & derivatives , Bradykinin/biosynthesis , Bradykinin/genetics , Bradykinin/pharmacology , Bradykinin B1 Receptor Antagonists , Bradykinin B2 Receptor Antagonists , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , Cell Adhesion/drug effects , Cell Adhesion Molecules/genetics , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Endothelium, Vascular/pathology , Female , Gene Expression Regulation/drug effects , Hyaluronan Receptors/biosynthesis , Hyaluronan Receptors/genetics , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , L-Selectin/biosynthesis , L-Selectin/genetics , Leukocytes/metabolism , Male , Oligopeptides/pharmacology , Peptidoglycan/toxicity , Prekallikrein/analysis , Rats , Rats, Inbred Lew , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/physiology , Specific Pathogen-Free Organisms , Synovial Membrane/blood supply , Synovial Membrane/metabolism , Synovial Membrane/pathology
4.
J Thromb Haemost ; 1(8): 1791-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12911595

ABSTRACT

We previously localized the heparin binding region on high molecular weight kininogen to domain 5 (D5) by quantifying the binding using surface plasmon resonance of D5 fused at its N-terminal to glutathione-S-transferase. We further examined GST-(H475-S626) which at 100 nm was previously shown to be ineffective in reversing the heparin acceleration of antithrombin inhibition of thrombin. However, we now show that at a concentration of 400 nm, complete reversal of accelerated inhibition occurred. To characterize the interacting sequences on D5, four peptides representing surface loops of a molecular model were synthesized. Peptides H475-H485 and G440-G455, rich in histidine and low in lysine, showed weak or no detectable binding in the absence of Zn++, but tighter binding in the presence of Zn++. H483-K497 containing three histidines and six lysines showed tight binding without Zn++, and increased in avidity with Zn++. In contrast, G486-K502, low in histidine and high in lysine, showed tight binding (KD = 0.8 microm) in the absence and presence of Zn++. Both H483-K497 and G486-K502 were effective in neutralizing the accelerated inhibition by heparin of thrombin by antithrombin in the absence of Zn++. Therefore, a set of lysine residues in the sequence of K487-K502 is responsible for Zn++-independent binding of heparin. Further, a group of histidine residues in sequence range of H475-H485 contributes to Zn++-dependent binding of heparin to HK-D5.


Subject(s)
Heparin/chemistry , Kininogen, High-Molecular-Weight/chemistry , Zinc/chemistry , Amino Acid Sequence , Antithrombins/chemistry , Biotinylation , Dose-Response Relationship, Drug , Edetic Acid/pharmacology , Gene Deletion , Glutathione Transferase/metabolism , Histidine/chemistry , Humans , Kinetics , Kininogen, High-Molecular-Weight/genetics , Lysine/chemistry , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Surface Plasmon Resonance , Thrombin/chemistry , Time Factors
5.
J Thromb Haemost ; 1(1): 164-70, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12871554

ABSTRACT

Previously we demonstrated that domain 5 (D5) of high-molecular-weight kininogen (HK) inhibits neovascularization in the chicken chorioallantoic membrane (CAM) assay and further found that kallikrein cleaved HK (HKa) inhibited FGF2-and VEGF-induced neovascularization, and thus was antiangiogenic. In this study, we sought to demonstrate whether uncleaved HK stimulates neovascularization and thus is proangiogenic. The chick chorioallantoic membrane was used as an in ovo assay of angiogenesis. Low-molecular-weight kininogen stimulates angiogenesis, indicating that D5 is not involved. Bradykinin stimulates neovascularization equally to HK and LK and is likely to be responsible for the effect of HK. A murine monoclonal antibody to HK (C11C1) also recognizes a similar component in chicken plasma as detected by surface plasmon resonance. Angiogenesis induced by FGF2 and VEGF is inhibited by this monoclonal antibody and is a more potent inhibitor of neovascularization induced by VEGF than an integrin alphavbeta3 antibody (LM 609). Our postulate that C11C1 inhibits the stimulation of angiogenesis by HK was confirmed when either C11C1 or D5 completely inhibited angiogenesis in the CAM induced by HK. Growth of human fibrosarcoma (HT-1080) on the CAM was inhibited by GST-D5 and C11C1. These results indicate HK is proangiogenic probably by releasing bradykinin and that a monoclonal antibody directed to HK could serve as an antiangiogenic agent with a potential for inhibiting tumor angiogenesis and other angiogenesis-mediated disorders.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Kininogen, High-Molecular-Weight/antagonists & inhibitors , Neovascularization, Physiologic/immunology , Allantois/blood supply , Animals , Antibodies, Monoclonal/immunology , Bradykinin/pharmacology , Chick Embryo , Chorion/blood supply , Dose-Response Relationship, Drug , Endothelial Growth Factors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Fibrosarcoma/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Kininogen, High-Molecular-Weight/immunology , Kininogen, High-Molecular-Weight/pharmacology , Kininogen, Low-Molecular-Weight/pharmacology , Lymphokines/pharmacology , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Surface Plasmon Resonance , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
FASEB J ; 15(13): 2365-76, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11689462

ABSTRACT

Proteolytic cleavage of single-chain, high molecular weight kininogen (HK) by kallikrein releases the short-lived vasodilator bradykinin and leaves behind a two-chain, high molecular weight kininogen (HKa) reported to bind to the beta2-integrin Mac-1 (CR3, CD11b/CD18, alphaMbeta2) on neutrophils and exert antiadhesive properties by binding to the urokinase receptor (uPAR) and vitronectin. We define the molecular mechanisms for the antiadhesive effects of HK related to disruption of beta2-integrin-mediated cellular interactions in vitro and in vivo. In a purified system, HK and HKa inhibited the binding of soluble fibrinogen and ICAM-1 to immobilized Mac-1, but not the binding of ICAM-1 to immobilized LFA-1 (CD11a/CD18, alphaLbeta2). This inhibitory effect could be attributed to HK domain 5 and to a lesser degree to HK domain 3, consistent with the requirement of both domains for binding to Mac-1. Accordingly, HK, HKa, and domain 5 inhibited the adhesion of Mac-1 but not LFA-1-transfected K562 human erythroleukemic cells to ICAM-1. Moreover, adhesion of human monocytic cells to fibrinogen and to human endothelial cells was blocked by HK, HKa, and domain 5. By using peptides derived from HK domain 5, the sequences including amino acids H475-G497 (and to a lesser extent, G440-H455) were identified as responsible for the antiadhesive effect, which was independent of uPAR. Finally, administration of domain 5 into mice, followed by induction of thioglycollate-provoked peritonitis, decreased the recruitment of neutrophils by approximately 70% in this model of acute inflammation. Taken together, HKa (and particularly domain 5) specifically interacts with Mac-1 but not with LFA-1, thereby blocking Mac-1-dependent leukocyte adhesion to fibrinogen and endothelial cells in vitro and in vivo and serving as a novel endogenous regulator of leukocyte recruitment into the inflamed tissue.


Subject(s)
Kininogen, High-Molecular-Weight/pharmacology , Leukocytes/drug effects , Peptides/pharmacology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Binding, Competitive/drug effects , Cell Adhesion/drug effects , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fibrinogen/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , K562 Cells , Kininogen, High-Molecular-Weight/chemistry , Leukocytes/cytology , Leukocytes/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/metabolism , Mice , Mice, Inbred Strains , Molecular Sequence Data , Neutrophil Infiltration/drug effects , Peritonitis/chemically induced , Peritonitis/pathology , Peritonitis/prevention & control , Plasmids/genetics , Thioglycolates/administration & dosage , U937 Cells
7.
Blood ; 96(2): 514-22, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10887113

ABSTRACT

Proteolytic cleavage of single-chain high molecular weight kininogen (HK) by kallikrein releases the short-lived vasodilator bradykinin and leaves behind 2-chain high molecular weight kininogen (HKa) that has been previously reported to exert antiadhesive properties as well as to bind to the urokinase receptor (uPAR) on endothelial cells. In this study we defined the molecular mechanisms for the antiadhesive effects of HKa related to disruption of integrin- and uPAR-mediated cellular interactions. Vitronectin (VN) but not fibrinogen or fibronectin-dependent alphavbeta(3) integrin-mediated adhesion of endothelial cells was blocked by HKa or its isolated domain 5. In a purified system, HKa but not HK competed for the interaction of VN with alphavbeta(3) integrin, because HKa and the isolated domain 5 but not HK bound to both multimeric and native VN in a Zn(2+)-dependent manner. The interaction between HKa or domain 5 with VN was prevented by heparin, plasminogen activator inhibitor-1, and a recombinant glutathione-S-transferase (GST)-fusion peptide GST-VN (1-77) consisting of the amino terminal portion of VN (amino acids 1-77), but not by a cyclic arginyl-glycyl-aspartyl peptide, indicating that HKa interacts with the amino terminal portion of VN ("somatomedin B region"). Furthermore, we have confirmed that HKa but not HK bound to uPAR and to the truncated 2-domain form of uPAR lacking domain 1 in a Zn(2+)-dependent manner. Through these interactions, HKa or its recombinant His-Gly-Lys-rich domain 5 completely inhibited the uPAR-dependent adhesion of myelomonocytic U937 cells and uPAR-transfected BAF-3 cells to VN and thereby promoted cell detachment. By immunogold electron microscopy, both VN and HK/HKa were found to be colocalized in sections from human atherosclerotic coronary artery, indicating that the described interactions are likely to take place in vivo. Taken together, HK and HKa inhibit different VN-responsive adhesion receptor systems and may thereby influence endothelial cell- or leukocyte-related interactions in the vasculature, particularly under inflammatory conditions. (Blood. 2000;96:514-522)


Subject(s)
Cell Adhesion , Kininogens/physiology , Receptors, Cell Surface/physiology , Receptors, Vitronectin/physiology , Arteriosclerosis/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fibrinogen/pharmacology , Humans , Kininogens/analysis , Kininogens/metabolism , Leukocytes/physiology , Molecular Weight , Monocytes , Osteosarcoma , Plasminogen Activator Inhibitor 1/analysis , Receptors, Cell Surface/analysis , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured , U937 Cells , Vitronectin/analysis , Vitronectin/metabolism , Vitronectin/pharmacology , Zinc/pharmacology
8.
Biochemistry ; 39(17): 5104-10, 2000 May 02.
Article in English | MEDLINE | ID: mdl-10819977

ABSTRACT

Previous investigations have shown that HK and its light chain bind heparin, preventing the enhancement of antithrombin inhibition of thrombin and potentiating the inhibition of plasma kallikrein by antithrombin. We found that both HK and HKa bound heparin, but HK exhibited a greater affinity. We therefore localized the binding sites for heparin on HK. HK domains 5 and 6 of the light chain as well as domain 3 from the heavy chain, expressed as glutathione S-transferase (GST) fusion proteins in Escherichia coli, were tested for binding to immobilized heparin by surface plasmon resonance using a BiaCore 2000 instrument. GST-D5, but not GST-D3, GST-D6, or GST, bound to heparin when the recombinant domains were present at a concentration of 70 nM. To localize more precisely the amino acid sequences on D5, both of the subdomains, histidine-glycine-rich GST-(K420-D474) and histidine-glycine-lysine-rich GST-(H475-S626), were expressed and tested for binding to immobilized heparin. The K(d) was much lower for GST-(K420-D474) than for GST-(H475-S626) in the presence or absence of Zn(2+). GST-(K420-D474) was effective in decreasing the rate of inactivation of thrombin by antithrombin in the presence of heparin and Zn(2+), while GST-(H475-S626) had no effect. We conclude that the binding of heparin to HK is a complex function of Zn(2+) interacting with histidines in the sequence K420-D474 to create high-affinity binding sites. HK has the potential to be an important modulator of heparin therapy.


Subject(s)
Heparin/chemistry , Kininogens/chemistry , Amino Acid Sequence , Binding Sites , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Heparin/metabolism , Kinetics , Kininogens/metabolism , Molecular Sequence Data , Protein Binding , Zinc
9.
J Biol Chem ; 275(30): 22756-63, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10801853

ABSTRACT

Factor XII deficiency has been postulated to be a risk factor for thrombosis suggesting that factor XII is an antithrombotic protein. The biochemical mechanism leading to this clinical observation is unknown. We have previously reported high molecular weight kininogen (HK) inhibition of thrombin-induced platelet aggregation by binding to the platelet glycoprotein (GP) Ib-IX-V complex. Although factor XII will bind to the intact platelet through GP Ibalpha (glycocalicin) without activation, we now report that factor XIIa (0. 37 microm), but not factor XII zymogen, is required for the inhibition of thrombin-induced platelet aggregation. Factor XIIa had no significant effect on SFLLRN-induced platelet aggregation. Moreover, an antibody to the thrombin site on protease-activated receptor-1 failed to block factor XII binding to platelets. Inhibition of thrombin-induced platelet aggregation was demonstrated with factor XIIa but not with factor XII zymogen or factor XIIf, indicating that the conformational exposure of the heavy chain following proteolytic activation is required for inhibition. However, inactivation of the catalytic activity of factor XIIa did not affect the inhibition of thrombin-induced platelet aggregation. Factor XII showed displacement of biotin-labeled HK (30 nm) binding to gel-filtered platelets and, at concentrations of 50 nm, was able to block 50% of the HK binding, suggesting involvement of the GP Ib complex. Antibodies to GP Ib and GP IX, which inhibited HK binding to platelets, did not block factor XII binding. However, using a biosensor, which monitors protein-protein interactions, both HK and factor XII bind to GP Ibalpha. Factor XII may serve to regulate thrombin binding to the GP Ib receptor by co-localizing with HK, to control the extent of platelet aggregation in vivo.


Subject(s)
Factor XII/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombin/antagonists & inhibitors , Amino Acid Sequence , Antibodies/immunology , Binding, Competitive , Blotting, Western , Humans , Platelet Glycoprotein GPIb-IX Complex/immunology , Protein Binding , Receptor, PAR-1 , Receptors, Thrombin/metabolism , Thrombin/pharmacology
10.
J Clin Invest ; 100(6): 1481-7, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9294114

ABSTRACT

The urokinase receptor (uPAR) binds urokinase-type plasminogen activator (u-PA) through specific interactions with uPAR domain 1, and vitronectin through interactions with a site within uPAR domains 2 and 3. These interactions promote the expression of cell surface plasminogen activator activity and cellular adhesion to vitronectin, respectively. High molecular weight kininogen (HK) also stimulates the expression of cell surface plasminogen activator activity through its ability to serve as an acquired receptor for prekallikrein, which, after its activation, may directly activate prourokinase. Here, we report that binding of the cleaved form of HK (HKa) to human umbilical vein endothelial cells (HUVEC) is mediated through zinc-dependent interactions with uPAR. These occur through a site within uPAR domains 2 and 3, since the binding of 125I-HKa to HUVEC is inhibited by vitronectin, anti-uPAR domain 2 and 3 antibodies and soluble, recombinant uPAR (suPAR), but not by antibody 7E3, which recognizes the beta chain of the endothelial cell vitronectin receptor (integrin alphavbeta3), or fibrinogen, another alphavbeta3 ligand. We also demonstrate the formation of a zinc-dependent complex between suPAR and HKa. Interactions of HKa with endothelial cell uPAR may underlie its ability to promote kallikrein-dependent cell surface plasmin generation, and also explain, in part, its antiadhesive properties.


Subject(s)
Endothelium, Vascular/metabolism , Kininogens/metabolism , Receptors, Cell Surface/metabolism , Cells, Cultured , Chromatography, Gel , Humans , Phosphatidylinositol Diacylglycerol-Lyase , Plasminogen Activators/metabolism , Prekallikrein , Receptors, Urokinase Plasminogen Activator , Receptors, Vitronectin/metabolism , Recombinant Proteins , Type C Phospholipases/pharmacology , Urokinase-Type Plasminogen Activator/metabolism , Vitronectin/metabolism , Vitronectin/pharmacology
11.
Blood ; 87(6): 2337-44, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8630396

ABSTRACT

In previous studies, we have shown that administration of monoclonal antibody (MoAb) C6B7 against human factor XII to baboons challenged with a lethal dose of Escherichia coli abrogates activation of the contact system and modulates secondary hypotension. To evaluate the contribution of activated contact proteases to the appearance of other inflammatory mediators in this experimental model of sepsis, we studied the effect of administration of MoAb C6B7 on activation of complement and fibrinolytic cascades, stimulation of neutrophil degranulation, and release of the proinflammatory cytokines, tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6). Activation of the complement system, as reflected by circulating C3b/c and C4b/c levels, was significantly reduced in five animals that had received MoAb C6B7 before a lethal dose of E coli as compared with five control animals that had been given a lethal challenge only. Inhibition of contact activation also modulated the fibrinolytic response, since the release of tissue-type plasminogen activator (t-PA) and the appearance of plasmin-alpha2-antiplasmin (PAP) complexes into the circulation was significantly attenuated upon pretreatment with anti-factor XII MoAb. In contrast, plasma levels of plasminogen activator inhibitor (PAI) were modestly enhanced in the treatment group. Degranulation of neutrophils, as assessed by circulating elastase-alpha1-protease inhibitor complexes, and release of IL-6 but not of TNF-alpha was decreased in anti-factor XII-treated animals. Observed differences in the inflammatory response between treatment and control groups were not likely due to different challenges, since the number of E coli that had been infused, as well as circulating levels of endotoxin after the challenge, were similar for both groups. These data suggest that activation of the contact system modulates directly or indirectly various mediator systems involved in the inflammatory response during severe sepsis in nonhuman primates.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bacteremia/blood , Complement Activation/drug effects , Escherichia coli Infections/blood , Factor XII/antagonists & inhibitors , Fibrinolysis/drug effects , Interleukin-6/metabolism , Pancreatic Elastase/metabolism , Shock, Septic/blood , Animals , Antibodies, Monoclonal/immunology , Bacteremia/therapy , Cytokines/metabolism , Escherichia coli Infections/therapy , Factor XII/immunology , Factor XII/physiology , Fibrinolysin/analysis , Immunization, Passive , Leukocyte Elastase , Mice , Neutrophils/enzymology , Neutrophils/metabolism , Papio , Plasminogen Activator Inhibitor 1/metabolism , Shock, Septic/therapy , Tissue Plasminogen Activator/metabolism , alpha-2-Antiplasmin/analysis
12.
Crit Care Med ; 23(1): 41-51, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7528118

ABSTRACT

OBJECTIVE: To test if serially sampled determinations of the contact system proteins and factor V have prognostic value for death in patients who develop the systemic inflammatory response syndrome. DESIGN: Prospective, observational study with sequential measurements in an inception cohort. SETTING: Medical intensive care unit (ICU) in a community hospital. PATIENTS: Over a 1-yr period, a population base sample of 23 patients was selected from all ICU admissions who met established criteria for the systemic inflammatory response syndrome. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Components of the contact system, factor XII, prekallikrein, high-molecular-weight kininogen, factor XI, alpha 2-macroglobulin-kallikrein complexes and factor V values were measured in plasma samples collected serially (day of admission, and at 2, 12, 24, 48 and/or 72 hrs or at discharge). Data were analyzed to determine if admission values or serially obtained values within 48 hrs were useful in predicting outcome. Fourteen patients survived and nine died. At admission, in all patients, assay values indicated that prekallikrein, high-molecular-weight kininogen, and factor V were significantly lower than normal (as observed in a range of 20 to 23 healthy adults), alpha 2-macroglobulin-kallikrein complexes were higher than normal, while concentrations of factor XII and factor XI were in the normal range. No differences were detected in the admission values between survivors and nonsurvivors, nor between patients with positive or negative blood cultures. However, subsequent values demonstrated a difference in values between survivors and nonsurvivors. Survivors showed improvement in high molecular weight kininogen values and higher than normal factor V values, as compared with nonsurvivors. CONCLUSIONS: Low or persistently low serial factor XII, high-molecular-weight kininogen and factor V values are associated with a poor prognosis, whereas high or increasing values of factor XII, high-molecular-weight kininogen, prekallikrein, and factor V all correlate with a favorable outcome.


Subject(s)
Factor V/analysis , Systemic Inflammatory Response Syndrome/blood , Aged , Blood Coagulation , Factor XI/analysis , Factor XII/analysis , Humans , Kininogens/blood , Prekallikrein/analysis , Prognosis , Prospective Studies , Survivors , Systemic Inflammatory Response Syndrome/mortality , alpha-Macroglobulins/analysis
13.
Blood ; 81(12): 3313-7, 1993 Jun 15.
Article in English | MEDLINE | ID: mdl-7685203

ABSTRACT

The objective of this study was to determine the role of the kallikrein-kinin system in healthy humans after intravenous administration of either Escherichia coli endotoxin or saline. We studied a total of 18 healthy nonsmoking volunteers, 23 to 38 years old, in an open-label study at the Critical Care Medicine Department, Clinical Center, National Institutes of Health (Bethesda, MD) in which some of the patients served as their own controls. After baseline data collection, the subjects received intravenously either E coli endotoxin (n = 15, 4 ng/kg of body weight) or saline (n = 8, controls). Signs, symptoms, systemic blood pressure, factor XII, plasma prekallikrein (PK), factor XI (FXI), antithrombin III (AT-III), high molecular weight kininogen (HK), and alpha 2-macroglobulin-kallikrein complexes were measured at baseline and 1, 2, 3, 5, and 24 hours after injection of either saline or endotoxin. After infusion of endotoxin, we found the functional plasma levels of FXI decreased at 2 hours (P < .05) and at 5 hours (P < .05). Functional PK was significantly depressed by 2 hours (P < .05), at 5 hours (P < .05), and at 24 hours (P < .01), whereas the PK antigen was only low at 5 hours (P < .05). These changes were accompanied by a significant increase in circulating alpha 2-macroglobulin-kallikrein complexes at 3 hours (P < .05) and 5 hours (P < .01). No significant changes occurred in the plasma levels of factor XII or HK. We concluded that clinical response to intravenous endotoxin in healthy human volunteers is associated with activation of the kallikrein-kinin systems. Further investigation is needed with specific inhibitors of the kallikrein-kinin system to define its primary or secondary role in the endotoxin-mediated reactions.


Subject(s)
Endotoxins/pharmacology , Escherichia coli , Kallikreins/metabolism , Kinins/metabolism , Adult , Antithrombin III/metabolism , Factor XI/metabolism , Humans , Kinetics , Kininogens/blood , Molecular Weight , Prekallikrein/metabolism , alpha-Macroglobulins/metabolism
15.
J Clin Invest ; 91(1): 61-8, 1993 Jan.
Article in English | MEDLINE | ID: mdl-7678610

ABSTRACT

The hypotension and disseminated intravascular coagulation (DIC) in bacteremia is thought to be mediated by the combined actions of cytokines, prostaglandins, and complement. The contact system, via the release of bradykinin and the activation of Factor XI, has been postulated to be contributing to the observed hypotension and DIC. Using a mAb to Factor XII (C6B7), we blocked the activation of the contact system in an established experimental baboon model in which Escherichia coli was infused to produce lethal bacteremia with hypotension. The untreated group (n = 5) displayed contact activation, manifested by a significant decrease in high molecular weight kininogen (HK) and a significant increase in alpha 2 macroglobulin-kallikrein complexes (alpha 2M-Kal). The C6B7-treated group (n = 5) showed an inactivation of Factor XII and the changes in HK and alpha 2M-Kal complexes were prevented. Both groups developed DIC manifested by a decrease in platelet, fibrinogen, and Factor V levels. The untreated group developed irreversible hypotension. The treated group experienced an initial hypotension that was reversed and extended the life of the animals. This study suggests that irreversible hypotension correlates with prolonged activation of the contact system, and specific antibody therapy can modulate both the pathophysiological and biochemical changes.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bacteremia/blood , Bacteremia/physiopathology , Disseminated Intravascular Coagulation/physiopathology , Escherichia coli Infections/blood , Escherichia coli Infections/physiopathology , Factor XII/physiology , Fibrinogen/metabolism , Hypotension/etiology , Kallikreins/metabolism , Kininogens/metabolism , alpha-Macroglobulins/metabolism , Animals , Blood Pressure , Factor XII/antagonists & inhibitors , Factor XII/immunology , Heart Rate , Humans , Kinetics , Papio , Platelet Count
16.
J Biol Chem ; 267(27): 19691-7, 1992 Sep 25.
Article in English | MEDLINE | ID: mdl-1527088

ABSTRACT

The first event leading to the activation of the plasma kallikrein-kinin system is the surface-dependent conversion of factor XII to an active enzyme. Factor XII autoactivation was investigated using dextran sulfate as a soluble activating surface, and the significance of aggregation and the nature of the conformational change were examined by ultraviolet difference spectroscopy, fluorescence and circular dichroism. Results indicate that DS500 (500-kDa dextran sulfate) induces aggregation of factor XII. Analysis of the binding data suggests that 165-192 factor XII molecules can bind to one DS500 chain, while a 1:1 stoichiometry is observed with 5-kDa dextran sulfate. The interaction of factor XII and dextran sulfate is a biphasic process. It is initiated by a fast contraction of the molecule upon binding, as revealed by an apparent increase in organized secondary structures, and then followed by a slow relaxation process during cleavage and subsequent activation. Overall, the results are consistent with a model in which factor XII undergoes conformational changes upon binding to the activating surface. The rapidity of autoactivation in the presence of DS500, as opposed to 5-kDa dextran sulfate, implies that aggregation provides a special mechanism whereby proteolytic cleavage is accomplished efficiently when factor XII molecules are bound side by side on the DS500 molecule.


Subject(s)
Dextran Sulfate/metabolism , Factor XII/metabolism , Circular Dichroism , Enzyme Activation , Factor XII/ultrastructure , Humans , In Vitro Techniques , Macromolecular Substances , Protein Binding , Protein Conformation , Scattering, Radiation , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet
17.
Am J Pathol ; 140(4): 897-906, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1373271

ABSTRACT

The hypotension in septicemia is believed to be mediated by the combined action of many mediators including cytokines, prostaglandins, and complement components. To evaluate the contribution of the contact/kinin-forming system to hypotension, the authors used an established experimental baboon model of bacteremia in which two concentrations of Escherichia Coli (E. coli) were used to produce lethal and nonlethal hypotension. The lethal group (n = 5) developed irreversible hypotension that significantly correlated with the decline in levels of high molecular weight kininogen (HK) and an increase in alpha 2 macroglobulin-kallikrein complexes (alpha 2M-kal). The nonlethal group (n = 9) experienced reversible hypotension, a less striking decline in HK, and only slight elevation in alpha 2M-kal. No significant changes were found in levels of factor XII, prekallikrein, and factor XI in either group. A significant change in the contact system, which reflects the fatal outcome, is the rise in alpha 2M-kal. This study suggests that irreversible hypotension correlates with prolonged activation of the contact system.


Subject(s)
Bacteremia/complications , Hypotension/complications , Animals , Bacteremia/mortality , Bacteremia/physiopathology , Blood Pressure , Escherichia coli , Hypotension/physiopathology , Kallikreins/blood , Kininogens/blood , Papio , alpha-Macroglobulins/analysis
18.
Thromb Haemost ; 66(5): 540-7, 1991 Nov 01.
Article in English | MEDLINE | ID: mdl-1725067

ABSTRACT

We examined in purified systems and in human plasma whether heparin serves as a contact system activating compound. Purified human factor XII zymogen was not activated by heparin through an autoactivation mechanism, but was activated in the presence of purified prekallikrein. Zn2+ (12 microM) did not support autoactivation by heparin. The activation of factor XII and the contact system by heparin in plasma anticoagulated with citrate or with hirudin (not chelating ions) was examined by the cleavage of 125I-labeled factor XII and high molecular weight kininogen (HK). Heparin at 1.6 and 16 USP U/ml was not able to produce activation, in contrast to dextran sulfate (20 micrograms/ml) which supported activation of both factor XII and HK. This study indicates that heparinized plasma does not support activation of the contact system mediated through activation of factor XII. It is not expected that heparin anticoagulant therapy will contribute to activation of the contact system.


Subject(s)
Factor XII/metabolism , Factor XIIa/metabolism , Heparin/pharmacology , Kininogens/metabolism , Complement C1 Inactivator Proteins/metabolism , Enzyme Activation , Factor XII/isolation & purification , Factor XII Deficiency/blood , Humans , Kallikreins/metabolism , Kinetics , Prekallikrein/pharmacology , Zinc/pharmacology , alpha-Macroglobulins/metabolism
19.
Blood ; 77(12): 2660-7, 1991 Jun 15.
Article in English | MEDLINE | ID: mdl-1710516

ABSTRACT

Activation of the contact system has been documented in severe sepsis and hereditary angioedema, but a sensitive, specific, and quantitative assay for assessing the degree of involvement of this proteolytic enzyme cascade is not yet available. We have developed a quantitative sandwich enzyme-linked immunosorbent assay (ELISA) for the alpha 2-macroglobulin-kallikrein (alpha 2M-Kal) complex using an F(ab')2 derivative of a monospecific polyclonal antibody against alpha 2 M as the capture antibody and a unique murine monoclonal antibody, 13G11, against the heavy chain of kallikrein as the detector antibody. The assay does not detect complexes in normal plasma but reacts with complexes generated by activating normal plasma with dextran sulfate at 4 degrees C in a range of 5 to 375 nmol/L. A close correlation of the ELISA with an amidolytic assay for alpha 2M-Kal was documented. Patients with sepsis syndrome but negative bacterial blood cultures did not show elevated plasma complexes, whereas a majority of those with positive blood cultures did show modest elevation and a single patient with septic shock showed a very high level of alpha 2M-Kal complex. Similarly, a patient with classic hereditary angioedema (HAE) showed increased concentration of complexes on three separate occasions during attacks but normal levels between attacks. Two other HAE patients did not show elevated levels at quiescent periods. The ELISA for alpha 2M-Kal appears to be sensitive, specific, and quantitative, and it can be used to reflect the degree of contact system activation in human sepsis and in HAE attacks.


Subject(s)
Angioedema/blood , Complement Activation , Kallikreins/metabolism , Sepsis/blood , alpha-Macroglobulins/metabolism , Antibodies, Monoclonal , Blotting, Western , Dextran Sulfate/pharmacology , Enzyme-Linked Immunosorbent Assay , Hexadimethrine Bromide , Humans , Prekallikrein/immunology , Shock, Septic/blood
20.
Am J Physiol ; 260(2 Pt 1): G213-9, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1996642

ABSTRACT

The plasma kallikrein-kinin system is activated in Gram-negative sepsis and typhoid fever, two diseases in which bacterial products have been shown to initiate inflammation. Because a single intraperitoneal injection of bacterial cell wall peptidoglycan-polysaccharide polymers from group A steptococci (PG-APS) into a Lewis rat produces a syndrome of relapsing polyarthritis and anemia, we investigated changes in the role of the kallikrein-kinin system in this model of inflammation. Coagulation studies after injection of PG-APS revealed an immediate and persistent decrease in prekallikrein levels. High-molecular-weight kininogen levels decreased significantly during the acute phase and correlated with the severity of arthritis. Factor XI levels were decreased only during the acute phase. Antithrombin III levels remained unchanged, indicating that neither decreased hepatic synthesis nor disseminated intravascular coagulation caused the decreased plasma contact factors. Plasma T-kininogen (an acute phase protein) was significantly elevated during the chronic phase. PG-APS failed to activate the contact system in vitro. Thus the kallikrein-kinin system plays an important role in this experimental model of inflammation, suggesting that activation of this system may play a role in the pathogenesis of inflammatory bowel disease and rheumatoid arthritis in which bacterial products might be etiologically important.


Subject(s)
Inflammation/etiology , Kallikrein-Kinin System/physiology , Streptococcal Infections/physiopathology , Streptococcus pyogenes/pathogenicity , Animals , Antithrombin III/metabolism , Cell Wall , Factor XI/analysis , Female , Kininogens/blood , Peptidoglycan/administration & dosage , Polysaccharides, Bacterial/administration & dosage , Prekallikrein/metabolism , Rats , Rats, Inbred Lew , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...