Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Biomolecules ; 10(6)2020 06 17.
Article in English | MEDLINE | ID: mdl-32560304

ABSTRACT

The blood coagulation factor XIII (FXIII) plays a critical role in supporting coagulation and fibrinolysis due to both the covalent crosslinking of fibrin polymers, rendering them resistant to plasmin lysis, and the crosslinking of fibrin to proteins of the fibrinolytic system. The hypochlorite-mediated oxidation of the blood coagulation factor XIII (FXIII) at the different stages of its enzymatic activation is studied for the first time in this paper. The consolidated results obtained with the aid of MS/MS, electrophoresis, and colorimetry demonstrate that in the process of FXIII's conversion into FXIIIa, the vulnerability of FXIII to hypochlorite-induced oxidation increased as follows: native FXIII < FXIII + Ca2+ << FXIII + Ca2+/thrombin. The modification sites were detected among all the structural regions of the catalytic FXIII-A subunit, except for the activation peptide, and embraced several sushi domains of the FXIII-B subunit. Oxidized amino acid residues belonging to FXIII-A are surface-exposed residues and can perform an antioxidant role. The regulatory FXIII-B subunits additionally contribute to the antioxidant defense of the catalytic center of the FXIII-A subunits. Taken together, the present data along with the data from previous studies demonstrate that the FXIII proenzyme structure is adapted to oxidation.


Subject(s)
Factor XIII/metabolism , Blood Coagulation , Factor XIII/chemistry , Factor XIII/isolation & purification , Female , Fibrinogen/chemistry , Fibrinogen/isolation & purification , Fibrinogen/metabolism , Humans , Male , Oxidation-Reduction
2.
Biomed Res Int ; 2017: 3571861, 2017.
Article in English | MEDLINE | ID: mdl-28894750

ABSTRACT

The role of factor XIII subunit A (FXIII-A) is not restricted to hemostasis. FXIII-A is also present intracellularly in several human cells and serves as a diagnostic marker in a wide range of dermatological diseases from inflammatory conditions to malignancies. In this review, we provide a guide on the still controversial interpretation of dermal cell types expressing FXIII-A and assess the previously described mechanisms behind their accumulation under physiological and pathological conditions of the human skin. We summarize the intracellular functions of FXIII-A as well as its possible sources in the extracellular space of the dermis with a focus on its relevance to skin homeostasis and disease pathogenesis. Finally, the potential role of FXIII-A in wound healing, as a field with long-term therapeutic implications, is also discussed.


Subject(s)
Factor XIII/genetics , Inflammation/diagnosis , Skin Diseases/diagnosis , Wound Healing/genetics , Factor XIII/isolation & purification , Hemostasis/genetics , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/therapy , Skin/metabolism , Skin/pathology , Skin Diseases/metabolism , Skin Diseases/pathology , Skin Diseases/therapy
3.
Dokl Biochem Biophys ; 467(1): 128-31, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27193716

ABSTRACT

For the first time, the induced oxidative modification of cellular fibrin-stabilizing factor (cFXIII) has been studied. According to the electrophoresis analysis, the conversion of oxidized cFXIII into FXIIIa resulted in producing the enzyme that significantly lost the initial enzymatic activity. At the same time, FXIIIa subjected to induced oxidation was completely devoid of enzymatic activity. The results of FTIR spectroscopy showed that the oxidation of cFXIII or FXIIIa was accompanied by profound changes both in chemical and spatial structures of the protein. The results of this study are in good agreement with our earlier assumption regarding the antioxidant role of the regulatory subunits B of plasma fibrin-stabilizing factor.


Subject(s)
Factor XIII/chemistry , Calcium/chemistry , Cations, Divalent/chemistry , Chromatography, Affinity , Electrophoresis, Polyacrylamide Gel , Factor XIII/isolation & purification , Female , Humans , Oxidants/chemistry , Oxidation-Reduction , Ozone/chemistry , Placenta/chemistry , Polymers/chemistry , Pregnancy , Protein Conformation , Spectroscopy, Fourier Transform Infrared
4.
J Biotechnol ; 168(4): 324-30, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24140601

ABSTRACT

The development of molecular probes is a prerequisite for activity-based protein profiling. This strategy helps in characterizing the catalytic activity and function of proteins, and how these proteins and protein complexes control biological processes of interest. These probes are composed of a reactive functional group and a reporter tag. The reactive group of these substrate probes has been considered to be important to their design, while the significance of the reporter tag is relatively underestimated. In this study we compare TAMRA-cadaverine and biotin-cadaverine, two substrate probes that have different reporter tags but an identical reactive functional group. We assess the on-chip transamidating activity of two transglutaminases; transglutaminase 2 and blood coagulation factor XIII. Activity assays were more easily executed when using the direct probe TAMRA-cadaverine. However the indirect probe, biotin-cadaverine, provided a wider dynamic range, higher signal-to-noise ratio, and lower limit of detection compared to TAMRA-cadaverine. Additionally, we successfully used the on-chip activity assay using the indirect probe to determine TG2 and FXIII activities in Hela cell lysates and human plasma samples, respectively. These results demonstrate that the reporter tag of the substrate probe is critical for protocol execution, sensitivity, and dynamic range of enzyme activity assays. Furthermore, this study provides a helpful guide for development of new probes, which is necessary for the identification of potential biomarkers and therapeutic targets for treating enzyme-related diseases.


Subject(s)
Cadaverine/chemistry , Factor XIII/isolation & purification , GTP-Binding Proteins/isolation & purification , Transglutaminases/isolation & purification , Biotin/chemistry , Factor XIII/chemistry , GTP-Binding Proteins/chemistry , HeLa Cells , Humans , Protein Glutamine gamma Glutamyltransferase 2 , Rhodamines/chemistry , Substrate Specificity , Transglutaminases/chemistry
6.
Transl Res ; 149(4): 187-95, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17383592

ABSTRACT

Fibrin-based sealants are commonly employed to arrest bleeding after surgery. Usually, fibrinogen obtained from pooled human plasma is used to prepare sealants, with attendant risk of blood-borne infections. Availability of autologous fibrinogen would eliminate this risk. To prepare autologous fibrin sealant, fibrinogen was precipitated from human plasma using protamine. Under optimal conditions (10-mg/mL protamine and 22 degrees C), 96 +/- 4% of clottable fibrinogen was recovered by a simple and inexpensive technique. Nearly 50% of the plasma factor XIII was also recovered with the fibrinogen. Using bovine thrombin, the fibrinogen was clotted (1) in a specially designed mold to measure tensile strength and (2) in a lap joint between 2 aortic vessel strips to measure adhesion strength. Tensile and adhesion strengths increased with increasing fibrinogen concentration, and they were increased by the addition of calcium chloride. The addition of aprotinin and -aminocaproic acid to the fibrinogen concentrate before clotting had no effect on the mechanical properties of the clots. After adding thrombin to sealant containing 15-mg/mL fibrinogen, maximum tensile strength was achieved in 1-5 min, and maximum adhesion strength was reached in 5-15 min. For the sealant with 30-60-mg/mL fibrinogen and added calcium, the tensile strength was equivalent to that of the commercial fibrin sealant Tisseel. The adhesion strength of sealant with 30-60-mg/mL fibrinogen exceeded the adhesive strength of Tisseel under identical conditions. Autologous fibrin sealant is an attractive alternative to commercial sealants. It can be readily prepared from 5-mL plasma or more and exhibits mechanical properties equivalent to those of the leading commercial sealant.


Subject(s)
Fibrin Tissue Adhesive/chemical synthesis , Adhesiveness , Anticoagulants/pharmacology , Antifibrinolytic Agents/pharmacology , Blood Coagulation/drug effects , Blood Proteins/drug effects , Blood Proteins/physiology , Calcium/pharmacology , Factor XIII/isolation & purification , Fibrinogen/isolation & purification , Heparin/pharmacology , Humans , Methods , Osmolar Concentration , Protamines/pharmacology , Temperature , Tensile Strength , Thrombin/pharmacology , Time Factors
7.
Diabetologia ; 49(5): 1071-80, 2006 May.
Article in English | MEDLINE | ID: mdl-16538489

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to determine the influence of type 2 diabetes on fibrinolysis by assessing interactions between the regulatory components of fibrinolysis and the fibrin clot, using fibrinogen purified from 150 patients with type 2 diabetes and 50 matched controls. METHODS: Clot lysis rates were determined by confocal microscopy. Plasmin generation was measured using a plasmin-specific chromogenic substrate. Surface plasmon resonance was used to determine the binding interactions between fibrin, tissue-type plasminogen activator (t-PA) and Glu-plasminogen; cross-linkage of plasmin inhibitor to fibrin by factor XIII was determined using a microtitre plate assay. RESULTS: Lysis of diabetic clots was significantly slower than that of controls (1.35 vs 2.92 microm/min, p<0.0001) and plasmin generation was significantly reduced. The equilibrium binding affinity between both t-PA and Glu-plasminogen and fibrin was reduced in diabetic subjects: t-PA, K (D)=0.91+/-0.3 micromol/l (control subjects), 1.21+/-0.5 micromol/l (diabetic subjects), p=0.001; Glu-plasminogen, K (D)=97+/-19 nmol/l (control subjects), 156+/-66 nmol/l (diabetic subjects), p=0.001. Cross-linkage of plasmin inhibitor to fibrin by factor XIII was enhanced in diabetic subjects, with the extent of in vitro cross-linkage correlating with in vivo glycaemic control (HbA(1c)) (r=0.59, p=0.001). CONCLUSIONS/INTERPRETATION: These results indicate that impairment of the fibrinolytic process in diabetic patients is mediated via a number of different mechanisms; these may be a consequence of post-translational modifications to fibrinogen molecules, resulting from their exposure to the abnormal metabolic milieu associated with diabetes.


Subject(s)
Diabetes Mellitus, Type 2/blood , Fibrin/pharmacology , Fibrinolysin/pharmacology , Hemolysis/drug effects , Aged , Blood Glucose/metabolism , Body Mass Index , Drug Resistance , Enzyme Activation , Factor XIII/isolation & purification , Female , Fibrinogen/chemistry , Fibrinogen/isolation & purification , Fibrinogen/metabolism , Glycated Hemoglobin/metabolism , Humans , Male , Mass Spectrometry , Middle Aged , Reference Values , Surface Plasmon Resonance
8.
J Thromb Haemost ; 3(11): 2487-96, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16241947

ABSTRACT

BACKGROUND: Factor (F)XIII B-subunit, which plays a carrier role for zymogen FXIIIA, is highly polymorphic, but the molecular basis for these polymorphisms and their relationship to disease remains unknown. OBJECTIVES: To screen the FXIIIB gene coding region for common variation and analyze possible functional effects. METHODS AND RESULTS: We examined the FXIIIB gene by PCR-SSCP and identified three common single nucleotide polymorphisms: A8259G, C29470T and A30899G. A8259G results in substitution of His95Arg in the second Sushi domain. An FXIII tetramer ELISA was developed to analyze B-subunit dissociation from A-subunit (leading to access to the catalytic site of FXIII). Increased subunit dissociation, 0.51 vs. 0.45 (fraction of total tetramer), was found in plasma from subjects possessing the Arg-allele. However, when the variants were purified to homogeneity and binding was analyzed by steady-state kinetics, no difference was observed. The relationship between His95Arg and venous thrombosis was investigated in 214 patients and 291 controls from Leeds. His/Arg + Arg/Arg genotypes were more frequent in patients than controls (22.4% vs. 15.1%). His95Arg was also investigated in the Leiden Thrombophilia Study, in which a similar difference was observed for 471 patients vs. 472 controls (18.5% vs. 14.0%), for a pooled odds ratio (OR) of 1.5 (CI95 1.1-2.0). CONCLUSIONS: We have identified three FXIIIB polymorphisms, one of which codes for substitution of His95Arg. The Arg95 variant associates with a moderately increased risk for venous thrombosis, and with increased dissociation of the FXIII subunits in plasma, although in vitro steady-state binding between purified subunits was not affected.


Subject(s)
Factor XIII/genetics , Polymorphism, Single Nucleotide , Thromboembolism/genetics , Venous Thrombosis/genetics , Binding Sites , Factor XIII/chemistry , Factor XIII/isolation & purification , Female , Gene Frequency , Genetic Testing , Genotype , Heterozygote , Homozygote , Humans , Kinetics , Male , Mutation , Phenotype , Protein Structure, Tertiary/genetics , Risk Factors , Thromboembolism/etiology , Venous Thrombosis/etiology
9.
Diabetologia ; 48(6): 1198-206, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15864538

ABSTRACT

AIMS/HYPOTHESIS: The precise mechanisms underlying the increased risk of cardiovascular disease (CVD) in type 2 diabetes are unclear. Fibrin clot structure has been related to CVD risk in the general population. We therefore assessed this in type 2 diabetic patients as a potential mechanism whereby diabetes influences CVD risk. METHODS: Fibrin clots were formed from fibrinogen purified from 150 subjects with type 2 diabetes and varying degrees of glycaemic control (assessed by HbA1c), and from 50 matched control subjects. Clot structure was assessed by turbidity, permeation and confocal microscopy. The specific effect of glucose itself was assessed by analysing the structure of clots formed from purified fibrinogen in the presence of increasing concentrations of the sugar. RESULTS: Clots formed by fibrinogen purified from type 2 diabetic subjects had a denser, less porous structure than those from control subjects. The structural changes found were related to the individual's glycaemic control; HbA1c correlated negatively with permeation coefficient (Ks) values (indicates clot pore size) (r = -0.57, p < 0.0001) and positively with maximum absorbance (indicator of fibre size) (r = 0.33, p < 0.0001), branch point number (r = 0.78, p < 0.0001) and fibre density (r = 0.63, p < 0.0001). The ambient glucose level influenced clot structure; hypo- (< 5 mmol) and hyperglycaemia (> or = 10 mmol/l) were both associated with a reduction in Ks values and maximum absorbance, and with increased fibre density and branch point number within clots. CONCLUSIONS/INTERPRETATION: The structural differences found to occur in type 2 diabetes and in association with hypo- and hyperglycaemia may confer increased resistance to fibrinolysis, and in consequence contribute to the increase in CVD risk in diabetic patients.


Subject(s)
Blood Coagulation , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Fibrin/chemistry , Fibrin/metabolism , Body Mass Index , Body Size , Cardiovascular Diseases/epidemiology , Diabetic Angiopathies/epidemiology , Factor XIII/isolation & purification , Factor XIII/metabolism , Female , Fibrin/drug effects , Fibrinogen/isolation & purification , Fibrinogen/metabolism , Humans , Hypoglycemic Agents/pharmacology , Lipids/blood , Male , Middle Aged , Reference Values , Risk Factors
10.
Semin Thromb Hemost ; 22(5): 427-36, 1996.
Article in English | MEDLINE | ID: mdl-8989827

ABSTRACT

With the increasing availability of human plasma this source was used to substitute for the production of factor XIII concentrate from placenta. Prior to changing the source material, the virus safety in accordance with the Committee for Proprietary Medicinal Products (CPMP) guidelines and the half-life were investigated. Concerning the virus safety, the following cumulative log 10 clearance factors were obtained: human immunodeficiency virus (HIV) > or = 18.9, herpes simplex virus (HSV-1) > or = 21.5, polio > or = 19.1, bovine viral diarrhea virus (BVDV) > or = 13.3. Half-life studies of factor XIII from plasma in comparison with factor XIII from placenta were done in rabbits by determination of the antigen and in patients with congenital factor XIII deficiency by determination of the activity and antigen. In rabbits, the terminal half-life of the antigen was 78.2 hours for factor XIII from placenta and 87.0 hours for factor XIII from plasma. In patients the half-lives were similar: 9.2 days for activity and antigen of factor XIII from plasma and 8.5 days (activity) versus 9.4 days (antigen) for the placenta-derived factor XIII. Some further clinical studies with factor XIII concentrates are also reviewed. Newer developments concerning recombinant factor XIII, its expression, characterization, and properties are summarized. Concerning the physicochemical data, the behavior in plasma was characterized by the formation of high-molecular-weight complexes, and first in vivo results, the recombinant factor XIII product was comparable to the naturally occurring material.


Subject(s)
Factor XIII Deficiency/drug therapy , Factor XIII/therapeutic use , Amino Acid Sequence , Animals , Cattle , Child , Clinical Trials as Topic , Drug Contamination , Factor XIII/genetics , Factor XIII/isolation & purification , Factor XIII/pharmacokinetics , Guidelines as Topic , Guinea Pigs , Half-Life , Humans , Molecular Sequence Data , Placenta/chemistry , Rabbits , Rats , Recombinant Proteins/therapeutic use , Safety , Virus Diseases/prevention & control , Virus Diseases/transmission , World Health Organization
11.
J Biol Chem ; 270(37): 21827-32, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7665605

ABSTRACT

The reaction of Factor XIIIa with fibrin is the last enzyme-catalyzed step on the coagulation cascade, leading to the formation of a normal blood clot. The finding that fibrin is preferred by the cross-linking enzyme about 10-fold over the circulating fibrinogen suggests the operation of a unique substrate-level control for the orderly functioning of the physiological process in the forward direction. An important task is to elucidate the molecular mechanism for the transmission of the signal generated by the thrombin-catalyzed cleavage in the central E domain of fibrin to the distant Factor XIIIa-reactive glutamine residues. By focusing on the substrate sites present in gamma chain remnants of D type domains of fibrinogen and by employing the approach of fragment complementation with the regulatory E domain, which represents the thrombin-modified portion of fibrin, we have now succeeded in reconstructing in solution the phenomenon of kinetic enhancement for the reaction with Factor XIIIa. Two D type preparations (truncated fibrinogen, approximately 250 kDa and D', approximately 105 kDa) were obtained by digestion of human fibrinogen with endo Lys-C. Neither product could be cross-linked by Factor XIIIa, but as shown by the incorporation of dansylcadaverine, both were acceptor substrates for the enzyme. The plasmin-derived D (approximately 105-kDa) product, however, could be cross-linked into DD dimers. In all cases, the admixture of E fragments exerted a remarkable boosting effect on the reactions with Factor XIIIa. Even with native fibrinogen as substrate, cross-linking of gamma chains was enhanced in the presence of E. Nondenaturing electrophoresis was used to demonstrate the complex forming potential of E fragments with fibrinogen, truncated fibrinogen, D', or D. The GPRP tetrapeptide mimic of the GPRV N-terminal sequence of the alpha chains in the E fragments, abolished both complex formation and the kinetic boosting effect of E on the reactions of substrates with Factor XIIIa. Thus, the N-terminal alpha chain sequences seem to act as organizing templates for spatially orienting the D domains, probably during the protofibrillar assembly of the fibrin units, for favorable reaction with Factor XIIIa.


Subject(s)
Fibrin/chemistry , Fibrin/metabolism , Fibrinogen/metabolism , Transglutaminases/chemistry , Transglutaminases/metabolism , Binding Sites , Cadaverine/analogs & derivatives , Electrophoresis, Polyacrylamide Gel , Factor XIII/isolation & purification , Fibrinogen/isolation & purification , Humans , Kinetics , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Substrate Specificity
12.
Thromb Haemost ; 74(2): 622-5, 1995 Aug.
Article in English | MEDLINE | ID: mdl-8584996

ABSTRACT

The pharmacokinetics and tolerability of factor XIII (FXIII) from plasma were compared with those of FXIII from placenta in a randomised, double-blind, crossover study involving 13 patients with congenital FXIII deficiency. Both FXIII activity and FXIII antigen were monitored. No difference was seen in the mean half-lives of the two preparations (9.3 days and 9.1 days for plasma and placenta FXIII activity, respectively). Response was similar for both preparations, but was slightly greater for FXIII from plasma (1.6 ormula: see text] vs 1.5 [formula: see text]). Similar results were found for recovery (65% vs 60%). The area under the data completed by extrapolation was significantly higher for FXIII from plasma. No differences between preparations in terms of efficacy or tolerability were observed. It can be concluded that treatment with FXIII concentrate from plasma is as efficient as with FXIII concentrate from placenta in terms of recovery and half-life. Both preparations were equivalent in terms of safety during the observation period. With the administration of monthly injections of approximately 30 U/kg serious bleeding events were prevented and no other serious adverse events occurred.


Subject(s)
Factor XIII Deficiency/therapy , Factor XIII/isolation & purification , Placenta/chemistry , Adolescent , Adult , Blood , Child , Cross-Over Studies , Double-Blind Method , Factor XIII/adverse effects , Factor XIII/pharmacokinetics , Female , Half-Life , Hemorrhage/prevention & control , Humans , Male , Middle Aged , Safety , Treatment Outcome
13.
J Biol Chem ; 269(45): 28414-9, 1994 Nov 11.
Article in English | MEDLINE | ID: mdl-7961782

ABSTRACT

In the presence of plasma transglutaminase (factor XIIIa) fibrin first undergoes intermolecular covalent cross-linking between its gamma chains to create gamma dimers followed by slower cross-linking among its alpha chains to form alpha polymers. Progressive cross-linking of gamma chain dimers occurs at the slowest rate, resulting in gamma trimers and gamma tetramers ("gamma multimers"). Most studies indicate that cross-linked fibrin clots become resistant to fibrinolysis, but the basis for this event is not clear. In this study, we explored the role of gamma chain multimerization compared with alpha polymerization as causal factors in time-dependent development of resistance to fibrinolysis. Fibrin clots prepared from native (intact) fibrinogen were incubated for up to 120 h at near physiological ionic strength and a factor XIIIa level approximating that in plasma. These clots were lysed by plasmin at rates that were inversely proportional to the level of gamma multimers, which increased progressively with the time of incubation. In contrast, fibrin cross-linked at high ionic strength (a condition under which only gamma dimers and alpha polymers form) or fibrin formed in the absence of factor XIII showed no time-dependent decrease in lysis rates. Fibrin cross-linked for a fixed time period with increasing amounts of factor XIIIa contained gamma multimer levels that were proportional to the factor XIIIa concentration and lysed at rates that were inversely proportional to the gamma multimer level. Furthermore, cross-linked fibrin formed from fibrinogen fraction I-9, which has limited potential for alpha polymerization, showed the same reduction in the lysis rate as native cross-linked fibrin. These findings indicate that development of resistance to fibrinolysis of cross-linked fibrin is not measurably dependent upon gamma dimer or alpha polymer formation but develops solely as a function of gamma multimerization.


Subject(s)
Cross-Linking Reagents , Fibrin/chemistry , Fibrin/metabolism , Fibrinolysis , Chromatography, Affinity , Factor XIII/isolation & purification , Factor XIII/metabolism , Fibrinogen/chemistry , Fibrinogen/isolation & purification , Fibrinogen/metabolism , Fibrinolysin/metabolism , Humans , Kinetics , Macromolecular Substances , Models, Structural , Plasminogen/isolation & purification , Plasminogen/metabolism , Protein Conformation , Time Factors , Transglutaminases/metabolism
14.
ASAIO J ; 40(3): M846-52, 1994.
Article in English | MEDLINE | ID: mdl-8555633

ABSTRACT

Monoprotein coatings of biomaterials with either natural adhesion molecules or genetically designed analogs have been used to facilitate attachment and spreading of endothelial cells. However, such treatments were found insufficient to maintain the integrity of the endothelial surface under turbulent flow conditions. In addition, when brought into contact with blood, these coatings were susceptible to plasma and cell proteinases that could readily destroy their structure and weaken cell adherence to the surface. In addressing these problems, we developed a cryoprecipitate-based coating that can firmly bind to any nonporous, prosthetic surface and interact with endothelial cells. The primary structure of the coating consisted of an autologous fibrin meshwork. It was refined by various compositions of the fibrinogen containing mixture and secured to polystyrene or polyurethane surfaces by dry-heat treatment. Further modulation of the coating was achieved by physically immobilizing various doses of heparin and insulin into the three dimensional matrix of the meshwork. Endothelial cells attached and grew much better on polyurethanes coated with this autologous protein complex than on a polystyrene tissue culture surface. With proper use of its capacity to mimic the properties of basal membrane, and absence of immunologic complications, the resulting coating may become an ideal multifunctional interface between cells and prosthetic materials.


Subject(s)
Biocompatible Materials , Blood Proteins/isolation & purification , Endothelium, Vascular/cytology , Animals , Cattle , Cell Adhesion , Cell Division , Cells, Cultured , Chemical Precipitation , Evaluation Studies as Topic , Factor XIII/isolation & purification , Fibrin , Fibrinogen/isolation & purification , Fibronectins/isolation & purification , Freezing , Humans , Surface Properties , Swine , Vitronectin/isolation & purification
15.
Anal Biochem ; 219(1): 43-8, 1994 May 15.
Article in English | MEDLINE | ID: mdl-8059954

ABSTRACT

Factor XIII A-chain-fibrin interactions regulate factor XIIIa formation and fibrin cross-linking. A microtiter plate assay was developed for studying these interactions. Microtiter plate wells were coated with fibrinogen and converted to fibrin by thrombin. After blocking the wells with bovine serum albumin, factor XIII A-chain was added and binding was monitored by incubating first with anti-factor XIII followed by anti-rabbit IgG-alkaline phosphatase. Enzymatic hydrolysis of p-nitrophenyl phosphate was quantitated by the absorbance at 405 nm. BInding was specific, sensitive, rapid, saturable, and reversible, requiring only nanograms of either factor XIII or fibrin. Binding was time- and concentration-dependent and independent of divalent cations. The bound material was identified as factor XIII A-chain by sodium dodecylsulfate-polyacrylamide gel electrophoresis and immunoblotting. Factor XIII binding was inhibited > 75% by 250 mM sodium chloride or 250 nM anti-factor XIII IgG. The method was also suitable for demonstrating binding using 0.8% plasma or with r-factor XIII expressed in Saccharomyces cerevisiae or Escherichia coli. This method is suitable for identifying the binding sites that are important for plasma factor XIII activation and factor XIIIa activity.


Subject(s)
Factor XIII/analysis , Factor XIII/metabolism , Fibrin/analysis , Fibrin/metabolism , Animals , Boron Compounds/pharmacology , Electrophoresis, Polyacrylamide Gel/methods , Factor XIII/isolation & purification , Fibrin/isolation & purification , Fibrinogen/metabolism , Goats/immunology , Humans , Immunoblotting/methods , Kinetics , Luminescent Measurements , Microchemistry , Molecular Weight , Oligopeptides/pharmacology , Protein Binding , Sensitivity and Specificity , Serum Albumin, Bovine , Sodium Chloride/pharmacology , Thrombin/antagonists & inhibitors , Thrombin/metabolism
16.
Protein Expr Purif ; 5(2): 125-32, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7914444

ABSTRACT

The purpose of this study was to develop an Escherichia coli expression system to facilitate study of the structure and function of blood coagulation factor XIII (FXIII) A-chains. We engineered an NcoI site into the full-length FXIII A-chain cDNA and subcloned it into pKK233-2 expression vector. A low level of full-length FXIII A-chain and a 30-kDa FXIII A-chain-related antigen were expressed in the JM 105 strain of E. coli. Protein sequencing of the 30-kDa protein demonstrated that it was synthesized by internal translation starting at either Met474 or Met475. We mutated the internal ribosome-binding sequences from AGGA to TGGT (pKF13A2 construct) and found that it yielded a 30-fold increase in the production of full-length FXIII A-chains. JM105 harboring pKF13A2 produced 20 mg of soluble FXIII A-chains antigen from 1 liter culture in TB medium. The recombinant FXIII A-chain was readily purified to homogeneity through PEG fractionation, Q-Sepharose, and mono-P column chromatography with a 2100-fold increase in specific activity and a yield of 150 to 200 micrograms of FXIII A-chains per liter of culture. The purified FXIII A-chains behaved as a dimer on gel filtration analysis, were thrombin- and calcium-activated, cross-linked fibrin, and bound to fibrin to the same extent as purified plasma FXIII A-chains and recombinant FXIII A-chains purified from yeast. These results document that FXIII A-chains can be readily expressed and purified from E. coli culture and that they retained properties similar to those of purified human factor XIII A-chains.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Factor XIII/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Base Sequence , Binding Sites , DNA, Complementary/genetics , Deoxyribonucleases, Type II Site-Specific , Escherichia coli , Factor XIII/genetics , Factor XIII/isolation & purification , Fibrin/metabolism , Gene Expression Regulation, Bacterial , Genetic Vectors , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Ribosomes/metabolism , Transglutaminases/metabolism
17.
Thromb Haemost ; 69(4): 397-400, 1993 Apr 01.
Article in English | MEDLINE | ID: mdl-8497854

ABSTRACT

To facilitate the analysis of the catalytic subunit of factor XIII we have developed a method for the immunoaffinity purification of this protein from platelets. This method employs a monoclonal antibody that binds to the a-subunit of factor XIII. The anti-factor XIII antibody was immobilized on agarose and then incubated with platelet lysate. Subsequently factor XIII was isolated from the platelet lysate in a single step with a 41% yield as measured by enzyme assay. The purified platelet factor XIII appeared nearly homogeneous when analyzed by polyacrylamide electrophoresis and by immunoblotting with another factor XIII monoclonal antibody.


Subject(s)
Antibodies, Monoclonal/immunology , Blood Platelets/chemistry , Chromatography, Affinity , Factor XIII/isolation & purification , Immunosorbent Techniques , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Factor XIII/immunology , Female , Humans , Mice , Mice, Inbred BALB C
20.
Int J Biochem ; 24(1): 91-7, 1992.
Article in English | MEDLINE | ID: mdl-1349868

ABSTRACT

1. FXIII was isolated and purified over 4000 fold from human placenta to apparent electrophoretic homogeneity by a new procedure including ethanol precipitation. DEAE-Cellulose, molecular sieving on Sephacryl S-300 and Phenyl-Sepharose chromatography. 2. Its pI was about 5.1. Under appropriate conditions, the incubation of FXIII in the presence of thrombin did not lead to inactivation cut in the polypeptidic chain. 3. FXIII was also activated by CaCl2 and, in a lesser extent, by other divalent cations like SrCl2, BaCl2 or MgCl2. 4. The binding of calcium to FXIII exhibited a negative cooperativity. 5. The activity-pH curve of the calcium-activated enzyme did not appear very different from that of the thrombin-activated enzyme.


Subject(s)
Factor XIII/metabolism , Placenta/metabolism , Calcium , Chromatography, Affinity , Chromatography, DEAE-Cellulose , Electrophoresis, Polyacrylamide Gel , Factor XIII/isolation & purification , Humans , Hydrogen-Ion Concentration , Isoelectric Focusing , Transglutaminases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...