Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
J Thromb Haemost ; 12(5): 606-13, 2014 May.
Article in English | MEDLINE | ID: mdl-24977287

ABSTRACT

BACKGROUND: High levels of activated protein­inhibitor complexes of the intrinsic coagulation proteins are associated with ischemic stroke (IS) but not with myocardial infarction (MI). This study was aimed at determining whether the antigen levels of coagulation factors(factor XII, FXII, and FXI and prekallikrein (PK)are associated with MI and IS, and whether this association is independent of levels of activated protein­inhibitor complexes. PATIENTS AND METHODS: The RATIO study included young women (< 50 years) with MI (N = 205)and IS (N = 175), and 638 healthy controls. Antigen levels of FXII, FXI and PK were measured and expressed as percentages of of those in pooled normal plasmas. Odds ratios (ORs) and corresponding 99% confidence intervals (CIs) were calculated for high levels (i.e. ≥ 90th percentile of controls) as measures of rate ratios. RESULTS: After adjustment for potential confounders, high levels of FXII antigen were not associated with MI risk or IS risk(OR(MI) 1.18, 99% CI 0.51­2.74; ORIS 1.03, 9% CI 0.41­2.55). High levels of FXI antigen were slightly associated with an increase in MI risk (OR(MI) 1.55, 9% CI 0.74­3.21), whereas there was a substantial association with IS risk (ORIS 2.65, 9% CI 1.27­5.56). PK antigen was slightly associated with MI risk but not with IS risk(ORMI 1.54, 9% CI 0.67­3.52; ORIS 0.90, 9% CI 0.35­2.33). All associations remained similar after adjustment for levels of protein­inhibitor complexes. CONCLUSION: Increased levels of FXI antigen were associated with an increase in IS risk, whereas they showed only a marginal association with MI risk. FXII antigen and PK antigen levels were not substantially associated with MI risk and IS risk.


Subject(s)
Antigens/immunology , Factor XII/immunology , Factor XI/immunology , Ischemia/blood , Myocardial Infarction/blood , Prekallikrein/immunology , Stroke/blood , Adolescent , Adult , Antigens/blood , Antigens/physiology , Case-Control Studies , Factor XI/physiology , Factor XII/physiology , Female , Fibrinolytic Agents/therapeutic use , Humans , Middle Aged , Odds Ratio , Prekallikrein/physiology , Risk Factors , Young Adult
2.
Adv Immunol ; 121: 41-89, 2014.
Article in English | MEDLINE | ID: mdl-24388213

ABSTRACT

Binding of negatively charged macromolecules to factor XII induces a conformational change such that it becomes a substrate for trace amounts of activated factor present in plasma (less than 0.01%). As activated factor XII (factor XIIa or factor XIIf) forms, it converts prekallikrein (PK) to kallikrein and kallikrein cleaves high molecular weight kininogen (HK) to release bradykinin. A far more rapid activation of the remaining unactivated factor XII occurs by enzymatic cleavage by kallikrein (kallikrein-feedback) and sequential cleavage yields two forms of activated factor XII; namely, factor XIIa followed by factor XII fragment (factor XIIf). PK circulates bound to HK and binding induces a conformational change in PK so that it acquires enzymatic activity and can stoichiometrically cleave HK to produce bradykinin. This reaction is prevented from occurring in plasma by the presence of C1 inhibitor (C1 INH). The same active site leads to autoactivation of the PK-HK complex to generate kallikrein if a phosphate containing buffer is used. Theoretically, formation of kallikrein by this factor XII-independent route can activate surface-bound factor XII to generate factor XIIa resulting in a marked increase in the rate of bradykinin formation as stoichiometric reactions are replaced by Michaelis-Menton, enzyme-substrate, kinetics. Zinc-dependent binding of the constituents of the bradykinin-forming cascade to the surface of endothelial cells is mediated by gC1qR and bimolecular complexes of gC1qR-cytokeratin 1 and cytokeratin 1-u-PAR (urokinase plasminogen activator receptor). Factor XII and HK compete for binding to free gC1qR (present in excess) while cytokeratin 1-u-PAR preferentially binds factor XII and gC1qR-cytokeratin 1 preferentially binds HK. Autoactivation of factor XII can be initiated as a result of binding to gC1qR but is prevented by C1 INH. Yet stoichiometric activation of PK-HK to yield kallikrein in the absence of factor XII can be initiated by heat shock protein 90 (HSP-90) which forms a zinc-dependent trimolecular complex by binding to HK. Thus, endothelial cell-dependent activation can be initiated by activation of factor XII or by activation of PK-HK. Hereditary angioedema (HAE), types I and II, are due to autosomal dominant mutations of the C1 INH gene. In type I disease, the level of C1 INH protein and function is proportionately low, while type II disease has a normal protein level but diminished function. There is trans-inhibition of the one normal gene so that functional levels are 30% or less and severe angioedema affecting peripheral structures, the gastrointestinal tract, and the larynx results. Prolonged incubation of plasma of HAE patients (but not normal controls) leads to bradykinin formation and conversion of PK to kallikrein which is reversed by reconstitution with C1 INH. The disorder can be treated by C1 INH replacement, inhibition of plasma kallikrein, or blockade at the bradykinin B-2 receptor. A recently described HAE with normal C1 INH (based on inhibition of activated C1s) presents similarly; the defect is not yet clear, however one-third of patients have a mutant factor XII gene. We have shown that this HAE has a defect in bradykinin overproduction whether the factor XII mutation is present or not, that patients' C1 INH is capable of inhibiting factor XIIa and kallikrein (and not just activated C1) but the functional level is approximately 40-60% of normal, and that α2 macroglobulin protein levels are normal. In vitro abnormalities can be suppressed by raising C1 INH to twice normal levels. Finally, aggregated proteins have been shown to activate the bradykinin-forming pathway by catalyzing factor XII autoactivation. Those include the amyloid ß protein of Alzheimer's disease and cryoglobulins. This may represent a new avenue for kinin-dependent research in human disease. In allergy (anaphylaxis; perhaps other mast cell-dependent reactions), the oversulfated proteoglycan of mast cells, liberated along with histamine, also catalyze factor XII autoactivation.


Subject(s)
Angioedemas, Hereditary/immunology , Angioedemas, Hereditary/pathology , Bradykinin/metabolism , Immunity, Innate , Inflammation Mediators/physiology , Signal Transduction/immunology , Angioedemas, Hereditary/etiology , Animals , Bradykinin/blood , Bradykinin/physiology , Complement C1 Inhibitor Protein/physiology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Factor XII/metabolism , Factor XII/physiology , Humans , Inflammation/blood , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/blood , Kininogen, High-Molecular-Weight/physiology , Prekallikrein/physiology
3.
Circulation ; 122(18): 1854-61, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20956210

ABSTRACT

BACKGROUND: Classically, intrinsic coagulation proteins are thought to have a minor role in hemostasis. Recently, these proteins, especially FXII, were implicated as possible key players in the pathogenesis of arterial thrombosis. This study aims to determine the risks of arterial thrombosis conferred by increased activation of intrinsic coagulation proteins in young women and the effect of oral contraceptive use on this association. METHODS AND RESULTS: The Risk of Arterial Thrombosis In relation to Oral contraceptives (RATIO) study is a population-based case-control study including young women (age 18 to 50 years) with myocardial infarction (n=205) and ischemic stroke (n=175) and 638 healthy controls. Intrinsic coagulation protein activation was determined by measuring activated protein-inhibitor complexes. This complex is with C1 esterase inhibitor (FXIIa-C1-INH, FXIa-C1-INH, Kallikrein-C1-INH) or antitrypsin inhibitor (FXIa-AT-INH). Odds ratios (ORs) and corresponding confidence intervals (95% CIs) were calculated with logistic regression. High levels of protein activation (>90th percentile of controls) showed an increased risk of ischemic stroke: FXIIa-C1-INH (OR, 2.1; 95% CI, 1.3 to 3.5), FXIa-C1-INH (OR, 2.8; 95% CI, 1.6 to 4.7), FXIa-AT-INH (OR, 2.3; 95% CI, 1.4 to 4.0), and Kallikrein-C1 (OR, 4.3; 95% CI, 2.6 to 7.2). If anything, myocardial infarction risk was only increased by Kallikrein-C1-INH (OR, 1.5; 95% CI, 0.9 to 2.5). Oral contraceptive use further increased the risks. CONCLUSIONS: High levels of activated proteins of the intrinsic coagulation system are associated with arterial thrombosis, whereas the strength of these associations differs for myocardial infarction and ischemic stroke. This contradicts similar analyses among men in the Northwick Park Heart Study. Together with the finding that oral contraceptive use further increases the risks, the question of whether the role of intrinsic coagulation proteins in the pathogenesis of arterial thrombosis is sex-specific is raised.


Subject(s)
Blood Coagulation Factors/physiology , Blood Coagulation/physiology , Contraceptives, Oral/adverse effects , Thrombosis/epidemiology , Thrombosis/physiopathology , Adolescent , Adult , Bradykinin/physiology , Case-Control Studies , Factor XI/physiology , Factor XII/physiology , Female , Humans , Kallikreins/physiology , Kininogens/physiology , Middle Aged , Myocardial Infarction/physiopathology , Prekallikrein/physiology , Risk Factors , Stroke/physiopathology , Young Adult
4.
Curr Drug Targets ; 6(5): 551-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16026275

ABSTRACT

Factor XII, plasma prekallikrein and high molecular weight kininogen were first identified as coagulation proteins in the intrinsic pathway because patients deficient in these proteins had marked prolongation of in vitro surface-activated coagulation time. However, deficiencies of these proteins are not associated with clinical bleeding. Paradoxically, studies suggest that these proteins have anticoagulant and profibrinolytic activities. In fact, association between deficiencies of these proteins and thrombosis has been reported. Also, deficiencies of these proteins, auto-antibodies to these proteins and anti-phospholipid antibodies are frequent hemostatis-related abnormalities found in unexplained recurrent aborters. Recently, evidence has accumulated for the presence of the kallikrein-kininogen-kinin system in the fetoplacental unit. Since contact proteins or kallikrein-kininogen-kinin system may play an important role in pregnancy especially in the fetoplacental unit, deficiencies of these proteins and/or auto-antibodies to these proteins may be associated with pregnancy losses. These possibilities will be reviewed, the functions of the individual components will be summarized, and their role in blood coagulation and pregnancy discussed.


Subject(s)
Factor XII/physiology , Kininogens/physiology , Pregnancy Complications/physiopathology , Prekallikrein/physiology , Animals , Female , Humans , Kininogen, High-Molecular-Weight/physiology , Plasma Kallikrein/physiology , Pregnancy , Pregnancy Complications, Hematologic/therapy
5.
Semin Thromb Hemost ; 30(1): 45-61, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15034797

ABSTRACT

We have demonstrated that domain 5 (D5, kininostatin) and cleaved high-molecular-weight kininogen (HKa) inhibit endothelial proliferation, migration, and neovascularization in the in ovo chicken chorioallantoic membrane (CAM) assay, and that D5 and HKa act by stimulating apoptosis and interfering with the cell cycle at the G (1)-S transition. Both intact high-molecular-weight kininogen (HK) and low-molecular-weight kininogen induce angiogenesis in the CAM assay by releasing bradykinin. A monoclonal antibody, mAb C11C1, targeted to HK D5, inhibits FGF2- (fibroblast growth factor-2) and vascular endothelial growth factor-stimulated angiogenesis in the CAM assay by interfering with the binding of HK to endothelial cells. We also demonstrate the inhibitory effects of both mAb C11C1 and glutathione-S-transferase-D5 on the growth of a human tumor supplied by CAM vessels.


Subject(s)
Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/etiology , Animals , Antibodies, Monoclonal/therapeutic use , Humans , Kininogens/physiology , Prekallikrein/physiology , Receptors, Cell Surface/physiology , Receptors, Urokinase Plasminogen Activator
6.
Int Immunopharmacol ; 2(13-14): 1841-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12489798

ABSTRACT

Understanding the importance and physiologic activity of the plasma kallikrein/kinin system (KKS) has been thwarted by the absence of an inclusive theory for its assembly and activation. The contact activation hypothesis describes the assembly and activation of this system in test tubes and disease states, but not under physiologic circumstances. Recent investigations have indicated a new cohesive hypothesis for understanding physiologic activation of this system. Prekallikrein (PK) and factor XI (FXI) through high molecular weight kininogen (HK) assemble on a co-localized, multiprotein receptor complex on endothelial cells that consists of at least cytokeratin 1 (CKI), gClqR, and urokinase plasminogen activator receptor (muPAR). When assembled on these proteins, prekallikrein becomes activated to kallikrein by the membrane-expressed enzyme prolylcarboxypeptidase (PRCP). Formed kallikrein then activates factor XII (FXII) for amplification of its activation and single chain urokinase. The plasma kallikrein/kinin system may serve as a physiologic counterbalance to the plasma renin angiotensin system (RAS) by lowering blood pressure and preventing thrombosis. Insights into the integrated role of these two systems may afford the development of novel therapeutic drugs to manage hypertension and thrombosis.


Subject(s)
Endothelium/physiology , Kallikrein-Kinin System/physiology , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase , Plasma Kallikrein/metabolism , Animals , Binding Sites , Binding, Competitive , Cell Membrane/metabolism , Cell Membrane/physiology , Endothelium/cytology , Endothelium/metabolism , Factor XI/metabolism , Factor XI/physiology , Factor XII/metabolism , Factor XII/physiology , Humans , Kallikrein-Kinin System/drug effects , Kininogen, High-Molecular-Weight/metabolism , Kininogen, High-Molecular-Weight/physiology , Plasma Kallikrein/physiology , Prekallikrein/metabolism , Prekallikrein/physiology
7.
Arch Pathol Lab Med ; 126(11): 1382-6, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12421145

ABSTRACT

OBJECTIVES: To review the literature for conditions, diseases, and disorders that affect activity of the contact factors, and further to review the literature for evidence that less than normal activity of any of the contact factors may be associated with thrombophilia. DATA SOURCES: MEDLINE search for English-language articles published from 1988 to 2001 and pertinent references contained therein, as well as search of references in recent relevant articles and reviews. STUDY SELECTION: Relevant clinical and laboratory information was extracted from selected articles. Meta-analysis was not feasible because of heterogeneity of reports. DATA EXTRACTION AND SYNTHESIS: Evidence for association of altered levels of the contact factors and thrombophilia was sought. A wide variety of disorders is associated with decreased activity of the contact factors; chief among these disorders are liver disease, hepatic immaturity of newborns, the antiphospholipid syndrome, and, for factor XII, being of Asian descent. These disorders are more common than homozygous deficiency. The few series and case reports of thrombophilic events in patients homozygous for deficiency of contact factors are not persuasive enough to support causality. The apparent association between levels consistent with heterozygosity (40%-60% of normal) of any of the contact factors (but especially factor XII) in persons with antiphospholipid antibodies appears to be due to falsely decreased in vitro activity levels of these factors, which are normal on antigenic testing. The apparent association with thrombosis is better explained by the antiphospholipid syndrome than by the modest reduction of the levels of contact factors. CONCLUSIONS: Presently, it is not recommended to measure activity of contact factors during routine evaluation of patients who have suffered venous or arterial thromboembolism or acute coronary syndromes.


Subject(s)
Factor XIa/physiology , Thrombophilia/diagnosis , Blood Coagulation/physiology , Blood Coagulation Tests , Factor XII/physiology , Factor XII Deficiency/diagnosis , Factor XII Deficiency/etiology , Factor XII Deficiency/physiopathology , Humans , Kininogen, High-Molecular-Weight/deficiency , Kininogen, High-Molecular-Weight/physiology , Practice Guidelines as Topic , Prekallikrein/deficiency , Prekallikrein/physiology , Thrombophilia/complications , Thrombophilia/physiopathology
8.
Sheng Li Ke Xue Jin Zhan ; 31(1): 7-12, 2000 Jan.
Article in Chinese | MEDLINE | ID: mdl-12532760

ABSTRACT

Current concept of blood coagulation is divided into two stages: an "initiation" stage which is handled by tissue factor pathway, and an "augmentation" stage handled by intrinsic pathway beginning in factor XI. Recent studies have demonstrated that the contact system is a modulator for vascular biology with vascular tone regulation, anticoagulant, profibrinolytic, antiadhesive and proinflammatory functions. Changes of contact system are associated with sepsis, thrombosis, etc.


Subject(s)
Blood Coagulation/physiology , Kininogen, High-Molecular-Weight/physiology , Prekallikrein/physiology , Animals , Factor XII/physiology , Factor XIIa/physiology , Humans , Thromboplastin/physiology
10.
Scand J Clin Lab Invest ; 59(1): 55-63, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10206098

ABSTRACT

With the present study, evidence is provided that prekallikrein (PK) in human plasma might be present in two different states, one of them removed along with IgG on Protein G columns. At a plasma dilution of 1 + 2.5, small amounts of an IgG fraction were left in plasma along with all of the PK. At a dilution of 1 + 11, nearly all IgG was removed. The removal in parallel of part of the PK was shown in immunoblot experiments and confirmed in amidase assays. One monoclonal antibody against PK (13G11) and two preparations of polyclonal antibodies were used for the immunoblot experiments. Different peptide substrates (S-2302, S-2222, Bz-Pro-Phe-Arg-pNA), along with protease inhibitors (soybean trypsin inhibitor, corn trypsin inhibitor, lima bean trypsin inhibitor) were used for the amidase assays. The amidase assays indicated that factors XII and XI were reduced by Protein G columns. In all experiments with extensive removal of IgG, protein recognized by the factor XII light chain mAb C6B7 was removed at the same time. This antibody preparation did not detect purified contact factors, but it did recognize a preparation of purified beta-FXIIa, and also significant amounts of protein present in plasma deficient in factor XII and not detectable in plasma deficient in PK. This protein accordingly seems to be connected with the PK fraction removed with IgG.


Subject(s)
Immunoglobulin G/isolation & purification , Prekallikrein/isolation & purification , Adult , Humans , Immunoblotting , Immunoglobulin G/blood , Male , Prekallikrein/physiology
13.
Thromb Haemost ; 78(1): 101-7, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9198136

ABSTRACT

In conclusion, a revised view of the contact system has been presented. This system has little to do with the initiation of hemostasis. Like lupus anticoagulants, deficiencies of contact proteins give prolonged APTTs but may be risk factors for thrombosis. BK from kininogens is a potent modulator of vascular biology inducing vasodilation, tissue plasminogen activator release, and prostacyclin liberation. Kininogens, themselves, are selective inhibitors of alpha-thrombin-induced platelet activation preventing alpha-thrombin from cleaving the cloned thrombin receptor after arginine41. Kininogens' alpha-thrombin inhibitory activity exists in intact kininogens, BK, and all of BK's breakdown products. HK also is the pivotal protein for contact protein assembly on endothelium. It is the receptor for prekallikrein which when bound to HK becomes activated to kallikrein by an endothelial cell enzyme system independent of activated forms of plasma factor XII. Prekallikrein activation on endothelial cells results in kinetically favorable single chain urokinase and plasminogen activation. Thus the "physiologic, negatively charged surface" for contact system activation is really the assembly of these proteins on cell membranes and activation by membrane-associated enzymes.


Subject(s)
Enzyme Precursors/physiology , Factor XII/physiology , Kininogens/physiology , Prekallikrein/physiology , Amino Acid Sequence , Cell Membrane/physiology , Humans , Kininogens/chemistry , Molecular Sequence Data , Molecular Weight , Structure-Activity Relationship
15.
Eur Heart J ; 14(6): 785-9, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8325306

ABSTRACT

Defective fibrinolysis may constitute a risk for the development of myocardial infarction in patients with ischaemic heart disease. We studied prospectively the factor XII-dependent plasminogen proactivator system in 49 survivors of an acute myocardial infarction. Blood samples were collected 8 weeks after hospital discharge. The factor XII-dependent fibrinolytic activity in the specimens was determined on fibrin plates after complete immuno-inhibition of the urokinase-like and the t-PA related fibrinolytic systems. During the subsequent follow-up period of 2.4 years, 10 patients developed recurrent myocardial infarction, whereas the remaining 39 patients did not. The reinfarction group of patients had a significantly lower median factor XII-dependent fibrinolytic activity (24.9 blood activating units (BAU).ml-1) than the patients without a relapse (41.9 BAU.ml-1, P < 0.02). Plasma concentrations of factor XII did not deviate significantly between the groups (P > 0.05), whereas the median plasma concentrations of prekallikrein was slightly lower in the reinfarction group (90%) than in the non-reinfarction group of patients (105%, P < 0.02). These observations point to an association between a depressed factor XII-dependent fibrinolytic activity and an enhanced risk of reinfarction in patients with a previous episode of acute myocardial infarction.


Subject(s)
Factor XII/physiology , Fibrinolysis/physiology , Myocardial Infarction/blood , Cholesterol/blood , Cholesterol, HDL/blood , Female , Humans , Male , Middle Aged , Myocardial Infarction/mortality , Prekallikrein/physiology , Prospective Studies , Recurrence , Risk Factors , Survival Rate , Thrombin/physiology , Triglycerides
16.
Agents Actions Suppl ; 38 ( Pt 2): 219-30, 1992.
Article in English | MEDLINE | ID: mdl-1462829

ABSTRACT

In recent years, extensive knowledge has been obtained on the structure/function relationships of blood coagulation proteins. In this overview, we present recent developments on the structure/function relationships of the contact activation proteins: factor XII, high molecular weight kininogen, prekallikrein, and factor XI, with the emphasis on the localization of domains on these proteins that are involved in the interaction with activators, substrates and cofactors.


Subject(s)
Blood Coagulation Factors/physiology , Amino Acid Sequence , Binding Sites , Blood Coagulation Factors/chemistry , Enzyme Activation , Factor XI/chemistry , Factor XI/physiology , Factor XII/chemistry , Factor XII/physiology , Humans , Kininogens/chemistry , Kininogens/physiology , Models, Structural , Molecular Sequence Data , Prekallikrein/chemistry , Prekallikrein/physiology , Protein Conformation
17.
Thromb Haemost ; 65(2): 144-8, 1991 Feb 12.
Article in English | MEDLINE | ID: mdl-1905069

ABSTRACT

An analysis was made of the various possible activators of single-chain urokinase-type plasminogen activator (scu-PA) in the dextran sulphate euglobulin fraction (DEF) of human plasma. scu-PA activators were detected in an assay system in which the substrate scu-PA, in physiological concentration (50 pM), was immuno-immobilized. After activation of the immobilized scu-PA for a certain period of time the activity of the generated amount of immuno-immobilized two-chain u-PA was determined with plasminogen and the chromogenic substrate S-2251. The scu-PA activator activity (scuPA-AA) in the DEF of plasmas deficient in factor XII or prekallikrein was about half of that in the DEF of normal plasma. Separation of scuPA-AA in the DEF by gel chromatography showed to major peaks, one eluting with an apparent Mr of 500,000 and the other around Mr 100,000. The former peak, which coincided with the activity peak of the kallikrein-kininogen complex, was absent in the DEF of plasma depleted of prekallikrein and therefore was identified as kallikrein. The latter peak was still present in the depleted plasma and most likely represents plasmin, because its scuPA-AA coincided with the activity peak of plasmin and could be fully inhibited by antibodies raised against human plasminogen. It is concluded that plasmin and the contact-activation factor kallikrein each contribute for about 50% to the scuPA-AA in the DEF. Compared on a molar basis, however, plasmin was found to be almost 1,000 times more effective than kallikrein, and we conclude, therefore, that in vivo plasmin is the primary activator of scu-PA and the role of the contact system is of secondary importance.


Subject(s)
Factor XII/physiology , Plasminogen Activators/metabolism , Prekallikrein/physiology , Serum Globulins/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Chemical Fractionation/methods , Chromatography, Gel , Dextran Sulfate , Humans
18.
Br J Urol ; 67(1): 83-7, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1993281

ABSTRACT

Urethral strictures induced by the use of latex catheters and combined silicone/latex catheters following open-heart surgery have been reported. These strictures differ from the post-catheterisation type in affecting a greater length of urethra. Recently it was shown that complement was activated by catheters which cause inflammation, in contrast to clinically silent catheters. The present study was designed to investigate a possible association between catheter-induced inflammation and activation of another mediator of inflammation, the contact system. We also investigated various in vitro conditions to optimise biocompatibility studies. Our data indicate that both the complement and the kallikrein-kinin system are activated by potentially harmful silicone/latex catheters and may be involved in the pathophysiology of catheter-induced urethral strictures. In vitro biocompatibility tests may be performed at both 20 and 37 degrees C. Glass test tubes may be used for in vitro complement but not for kallikrein-kinin investigations.


Subject(s)
Complement Activation/physiology , Kallikrein-Kinin System/physiology , Urinary Catheterization/adverse effects , Adult , Biocompatible Materials , Blood Coagulation/physiology , Female , Fibrinolysis/physiology , Humans , Kallikreins/blood , Latex , Male , Prekallikrein/physiology , Silicones , Temperature , Time Factors , Urethral Stricture/etiology
19.
Eur J Biochem ; 193(2): 495-9, 1990 Oct 24.
Article in English | MEDLINE | ID: mdl-2226467

ABSTRACT

In a system consisting of purified proteins inositol-phospholipid-accelerated activation of prekallikrein by alpha-factor XIIa was determined by measuring the appearance of kallikrein amidolytic activity towards the chromogenic substrate, H-D-Pro-Phe-Arg-NH-PhNO2 (PhNO2, 4-nitrophenyl). The activation reaction was ionic-strength dependent. In the absence of high-Mr kininogen optimal activity was recorded at I = 50 mM. Searching for conditions, which could change this optimum towards physiological values, high-Mr kininogen was added. This resulted in an inhibition of the activity, with no change in ionic strength optimum. If, however, Zn2+ were added concomitant with high-Mr kininogen, the inhibition was abolished and optimal activity recorded at physiological ionic strength. The optimal Zn2+ concentration was found to be 0.1 mM. Kinetic analysis of the reaction demonstrated that the kcat/Km was 1.2 x 10(5) M-1 s-1 in the absence and 1.1 x 10(6) M-1 s-1 in the presence of Zn2+. Zn2+ were also required for inositol-phospholipid-accelerated initiation of the contact activation in whole plasma.


Subject(s)
Blood Coagulation/physiology , Factor XIIa/physiology , Kininogens/physiology , Prekallikrein/physiology , Zinc/physiology , Cations, Divalent , Humans , Inositol/physiology , Kallikreins/metabolism , Kinetics , Oligopeptides/metabolism , Osmolar Concentration , Phospholipids/physiology
20.
Pharmacol Biochem Behav ; 35(2): 437-42, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2320652

ABSTRACT

Japanese healthy male subjects were divided into two groups, i.e., a normal aldehyde dehydrogenase (ALDH) group with a low Km isozyme of ALDH for acetaldehyde, and a deficient group without it. After intake of 0.4 g/kg alcohol, the deficient group showed high levels of blood acetaldehyde, facial flushing including an increased pulse rate and a fall in diastolic blood pressure, while the normal group did not manifest these changes. In the deficient group, the total kininogen concentration gradually decreased after alcohol intake due to a reduction in low molecular weight kininogen, and plasma prekallikrein remained unchanged. The normal group showed no significant changes in any of these values after alcohol intake. In an in vitro study with pooled plasma, the low concentrations of urinary kallikrein caused a decrease in the low molecular weight kininogen only. These results suggest that kinins released by acetaldehyde-induced activation of glandular kallikreins are associated with the changes in cardiovascular symptoms in deficient group which display flushing after alcohol intake.


Subject(s)
Alcohol Drinking/physiology , Aldehyde Dehydrogenase/deficiency , Flushing/etiology , Kininogens/physiology , Prekallikrein/physiology , Acetaldehyde/blood , Adolescent , Adult , Blood Pressure/drug effects , Ethanol/blood , Ethanol/pharmacology , Face , Flushing/physiopathology , Heart Rate/drug effects , Humans , Kininogens/blood , Male
SELECTION OF CITATIONS
SEARCH DETAIL