Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Methods Mol Biol ; 1100: 189-205, 2014.
Article in English | MEDLINE | ID: mdl-24218261

ABSTRACT

C1 inhibitor is a multipotent serpin capable of inhibiting the classical and the lectin pathways of complement, the fibrinolytic system, and contact/kinin system of coagulation. Deficiency of C1 inhibitor manifest as hereditary angioedema (HAE), an autosomal dominant hereditary disease. Measuring the C1 inhibitor level is of vital importance for the diagnosis of HAE and also for monitoring patients receiving C1 inhibitor for therapy. Determination of the antigenic C1 inhibitor level by the radial immunodiffusion (RID) technique is described in detail in this chapter. The presented purification method of plasma C1 inhibitor is primarily based on its high carbohydrate content and its affinity to the lectin jacalin.


Subject(s)
Complement C1 Inactivator Proteins/isolation & purification , Complement C1 Inactivator Proteins/metabolism , Chromatography, Affinity , Complement C1 Inactivator Proteins/immunology , Complement C1 Inhibitor Protein , Humans , Immunodiffusion/methods
2.
Amino Acids ; 44(5): 1381-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23512611

ABSTRACT

The protocol consists of running a native gel with in-gel digestion by proteases, subsequent mass spectrometrical determination of protein sequence and modifications, followed by electro-elution and conformational analysis using melting point and circular dichroism. Finally, the eluted protein is tested for preserved function. Herein, C1 esterase inhibitor is applied on a native gel; in-gel digestion by proteases is carried out and peptides are identified by nano-LC-ESI-CID/ETD-MS/MS using an ion trap for generation of peptide sequences and protein modifications. Protein from replicate bands from the same gel is electro-eluted and used for determination of the melting point and used for circular dichroism analysis. Additional bands from the native gel are either in-gel digested with asparaginase to generate deamidation or PNGase F for deglycosylation, followed by mass spectrometry, conformational and functional studies. Preserved conformation and function of the C1 esterase inhibitor was shown. This protocol can be completed in 1 week.


Subject(s)
Complement C1 Inactivator Proteins/chemistry , Chromatography, High Pressure Liquid , Circular Dichroism , Complement C1 Inactivator Proteins/isolation & purification , Complement C1 Inhibitor Protein , Humans , Molecular Weight , Native Polyacrylamide Gel Electrophoresis/methods , Protein Processing, Post-Translational , Protein Structure, Secondary , Proteins/chemistry , Proteins/isolation & purification , Sequence Analysis, Protein/methods , Spectrometry, Fluorescence , Tandem Mass Spectrometry , Transition Temperature
3.
Protein Expr Purif ; 88(2): 173-82, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23314348

ABSTRACT

MASP-1 is a protease of the lectin pathway of complement. It is homologous with MASP-2, previously thought both necessary and sufficient for lectin pathway activation. Recently MASP-1 has taken centre stage with the observation that it is crucial to the activation of MASP-2 and thus central to complement activation. Numerous additional functions have been suggested for MASP-1 and its importance is obvious. Yet, thorough analyses of proteolytic activities and physiological roles in the human scenario have been hampered by difficulties in purifying or producing full-length human MASP-1. We present the successful expression of full-length recombinant human MASP-1 entirely in the zymogen form in a mammalian expression system. We found that the catalytic activity of MASP-1 suppresses its expression through rapid auto-activation and auto-degradation. This auto-degradation was not inhibited by the addition of inhibitors to the culture medium, and it was subsequently found to occur intracellularly. Numerous mutations aimed at attenuating auto-activation or preventing auto-degradation failed to rescue expression, as did also attempts at stabilizing the protease by co-expression with MBL or ficolins or expression in hepatocyte cell lines, representing the natural site of synthesis. The active protease was finally produced through co-expression with the serine protease inhibitor C1 inhibitor. We demonstrate that the expressed protease is capable of binding MBL and auto-activating, and is catalytically active. We have generalized the concept to the expression also of MASP-2 entirely in its zymogen form and with improved yields. We suggest a general advantage of expressing aggressive, autocatalytic proteases with their cognate inhibitors.


Subject(s)
Cloning, Molecular , Complement C1 Inactivator Proteins/genetics , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Animals , Cell Line , Cloning, Molecular/methods , Complement C1 Inactivator Proteins/isolation & purification , Complement C1 Inactivator Proteins/metabolism , Enzyme Precursors/genetics , Enzyme Precursors/isolation & purification , Enzyme Precursors/metabolism , Gene Expression , Genetic Vectors/genetics , Hep G2 Cells , Humans , Lectins/genetics , Lectins/isolation & purification , Lectins/metabolism , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/isolation & purification , Mannose-Binding Lectin/metabolism , Mannose-Binding Protein-Associated Serine Proteases/isolation & purification , Mutagenesis, Site-Directed , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ficolins
4.
J Biotechnol ; 162(2-3): 319-26, 2012 Dec 31.
Article in English | MEDLINE | ID: mdl-22995741

ABSTRACT

C1 inhibitor (C1INH) is a single-chain glycoprotein that inhibits activation of the contact system of coagulation and the complement system. C1INH isolated from human blood plasma (pd-hC1INH) is used for the management of hereditary angioedema (HAE), a disease caused by heterozygous deficiency of C1INH, and is a promise for treatment of ischemia-reperfusion injuries like acute myocardial or cerebral infarction. To obtain large quantities of C1INH, recombinant human C1INH (rhC1INH) was expressed in the milk of transgenic rabbits (12 g/l) harboring genomic human C1INH sequences fused to 5' bovine αS(1) casein promoter sequences. Recombinant hC1INH was isolated from milk to a specific activity of 6.1 U/mg and a purity of 99%; by size-exclusion chromatography the 1% impurities consisted of multimers and N-terminal cleaved C1INH species. Mass spectrometric analysis of purified rhC1INH revealed a relative molecular mass (M(r)) of 67,200. Differences in M(r) on SDS PAGE and mass spectrometric analysis between rhC1INH and pd-hC1INH are explained by differential glycosylation (calculated carbohydrate contents of 21% and 28%, respectively), since protein sequencing analysis of rhC1INH revealed intact N- and C-termini. Host-related impurity analysis by ELISA revealed trace amounts of rabbit protein (approximately 10 ppm) in purified batches, but not endogenous rabbit C1INH. The kinetics of inhibition of the target proteases C1s, Factor XIIa, kallikrein and Factor XIa by rhC1INH and pd-hC1INH, indicated comparable inhibitory potency and specificity. Recently, rhC1INH (Ruconest(®)) has been approved by the European Medicines Agency for the treatment of acute attacks of HAE.


Subject(s)
Complement C1 Inactivator Proteins/chemistry , Genetic Engineering/methods , Milk/chemistry , Recombinant Proteins/chemistry , Animals , Animals, Genetically Modified , Cattle , Complement C1 Inactivator Proteins/biosynthesis , Complement C1 Inactivator Proteins/genetics , Complement C1 Inactivator Proteins/isolation & purification , Complement C1 Inhibitor Protein , Humans , Kinetics , Lactation , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
5.
BioDrugs ; 25(5): 317-27, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21942916

ABSTRACT

Intravenous nanofiltered human C1 inhibitor (C1-INH NF) concentrate (Cinryze®) is used as a direct replacement of deficient levels of plasma C1 inhibitor in patients with hereditary angioedema (HAE). In the EU, C1-INH NF concentrate 1000 U is indicated in the treatment, pre-procedural prevention, and routine prevention of angioedema attacks in adults and adolescents with HAE. Intravenous C1-INH NF concentrate 1000 U effectively relieved angioedema attacks in patients with HAE. In a randomized, double-blind trial in pediatric and adult patients, the median time to onset of unequivocal relief from an attack was significantly shorter with C1-INH NF concentrate than with placebo. In an open-label trial, both unequivocal relief and clinical relief were shown in the majority of attacks within 1 and 4 hours of infusion of C1-INH NF concentrate, regardless of the site (i.e. gastrointestinal, cutaneous, laryngeal, or genitourinary) of the defining symptom. When administered prior to a procedure, open-label intravenous C1-INH NF concentrate 1000 U reduced the incidence of angioedema attacks during and after a variety of dental, surgical, or interventional diagnostic procedures in pediatric and adult patients with HAE. Routine preventative treatment with intravenous C1-INH NF concentrate 1000 U every 3 or 4 days reduced the number of angioedema attacks. In a randomized, double-blind, crossover trial in pediatric and adult patients with HAE, the mean normalized number of attacks per 12-week period was significantly lower during routine prevention with C1-INH NF concentrate than with placebo. Routine prevention with C1-INH NF concentrate reduced the median monthly attack rate from baseline in an open-label trial. Intravenous C1-INH NF concentrate was well tolerated in clinical trials in patients with HAE. No cases of viral transmission were reported.


Subject(s)
Angioedemas, Hereditary/drug therapy , Complement C1 Inactivator Proteins/isolation & purification , Complement C1 Inactivator Proteins/pharmacokinetics , Complement C1 Inactivator Proteins/therapeutic use , Filtration/methods , Adult , Angioedemas, Hereditary/prevention & control , Child , Complement C1 Inactivator Proteins/administration & dosage , Complement C1 Inhibitor Protein , Double-Blind Method , Female , Humans , Infusions, Intravenous , Male , Nanotechnology/methods
6.
Biologicals ; 35(3): 173-81, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17071103

ABSTRACT

We studied the efficacy of virus reduction by three process steps (polyethylene glycol 4000 (PEG) precipitation, pasteurization, and 15nm virus filtration) in the manufacturing of C1-inhibitor NF. The potential prion removing capacity in this process was estimated based on data from the literature. Virus studies were performed using hepatitis A virus (HAV) and human immunodeficiency virus (HIV) as relevant viruses and bovine viral diarrhea virus (BVDV), canine parvovirus (CPV) and pseudorabies virus (PRV) as model viruses, respectively. In the PEG precipitation step, an average reduction in infectious titer of 4.5log(10) was obtained for all five viruses tested. Pasteurization resulted in reduction of infectious virus of >6log(10) for BVDV, HIV, and PRV; for HAV the reduction factor was limited to 2.8log(10) and for CPV it was zero. Virus filtration (15nm) reduced the infectious titer of all viruses by more than 4.5log(10). The overall virus reducing capacity was >16log(10) for the LE viruses. For the NLE viruses CPV and HAV, the overall virus reducing capacities were >8.7 and >10.5log(10), respectively. Based on literature and theoretical assumptions, the prion reducing capacity of the C1-inhibitor NF process was estimated to be >9log(10).


Subject(s)
Biological Products/isolation & purification , Complement C1 Inactivator Proteins/isolation & purification , Serpins/isolation & purification , Viruses/isolation & purification , Animals , Cattle , Cell Line , Chemical Precipitation , Complement C1 Inhibitor Protein , Diarrhea Viruses, Bovine Viral/isolation & purification , Disinfection , Dogs , Drug Contamination , Filtration , HIV/isolation & purification , Hepatitis A virus/isolation & purification , Herpesvirus 1, Suid/isolation & purification , Humans , Nanotechnology , Parvovirus, Canine/isolation & purification , Polyethylene Glycols , Safety , Swine
7.
Glycobiology ; 14(1): 51-64, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14514717

ABSTRACT

Human C1 inhibitor (hC1INH) is a therapeutic N, O-glycoprotein with a growing number of clinical applications, but the current natural supplies are not likely to meet the clinical demands. Therefore, recombinant approaches are of interest, whereby specific attention has to be paid to the generated glycosylation patterns. Here, the N,O-glycoprotein was expressed in the mammary gland of transgenic rabbits and subjected to glycan analysis. After release of the N-glycans of recombinant-rabbit human C1 inhibitor (rhC1INH) by peptide-N4-(N-acetyl-beta-glucosaminyl)asparagine amidase F, the oligosaccharides were separated from the O-glycoprotein by centrifugal filtration, then fractionated by a combination of anion-exchange, normal-phase, and high-pH anion-exchange liquid chromatography. The O-glycans, released from the O-glycoprotein by alkaline borohydride treatment, were fractionated by anion-exchange high-performance liquid chromatography (HPLC). The structures of individual components were analysed by 500 MHz 1H NMR spectroscopy, in most cases combined with MALDI-TOF MS. In contrast to the structural data reported for native serum hC1INH, rhC1INH contained a broad array of different N-glycans, made up of oligomannose-, hybrid-, and complex-type structures. In the case of complex-type N-glycans (partially) (alpha2-6)-sialylated (N-acetylneuraminic acid only), mono- and diantennary chains were found; part of the diantennary structures were (alpha1-6)-core-fucosylated or (alpha1-3)-fucosylated in the lower or upper antenna (Lewis x). The manno-oligosaccharide pattern of part of the hybrid- and oligomannose-type structures indicates that besides the usual N-glycan processing route, also the alternative endo-mannosidase pathway is followed. The small core 1-type O-glycans showed the usual (alpha2-3)- and (alpha2-6)-sialylation pattern of O-glycoproteins of nonmucinous origin.


Subject(s)
Complement C1 Inactivator Proteins/genetics , Milk/chemistry , Polysaccharides/chemistry , Recombinant Proteins/metabolism , Animals , Animals, Genetically Modified , Carbohydrate Conformation , Carbohydrate Sequence , Chromatography, High Pressure Liquid/methods , Complement C1 Inactivator Proteins/chemistry , Complement C1 Inactivator Proteins/isolation & purification , Female , Humans , Mammary Glands, Animal/physiology , Molecular Sequence Data , Oligosaccharides/chemistry , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
8.
Blood Coagul Fibrinolysis ; 13(8): 711-4, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12441910

ABSTRACT

C1-inhibitor is an important inhibitor of plasma kallikrein and C1, but also has inhibitory activity against numerous other plasma proteinases such as plasmin. The relevance of plasmin inhibition by the C1-inhibitor has been debated, with some evidence showing that plasmin causes significant proteolysis of C1-inhibitor. In the present study, we show that C1-inhibitor in its native state will inhibit plasmin without being significantly degraded, in a manner typical of all serpin reactions. However, if C1-inhibitor is in a denatured polymeric state (as can easily occur during storage, or as produced by heating of the native protein), it will be extensively degraded by plasmin. In addition, we show that hydrophobic interaction chromatography is an effective method to remove trace contaminants of inactive C1-inhibitor polymers.


Subject(s)
Blood Preservation/methods , Complement C1 Inactivator Proteins/metabolism , Fibrinolysin/antagonists & inhibitors , Chromatography , Complement C1 Inactivator Proteins/isolation & purification , Complement C1 Inhibitor Protein , Cysteine Proteinase Inhibitors/isolation & purification , Cysteine Proteinase Inhibitors/metabolism , Dimerization , Fibrinolysin/metabolism , Humans , Protein Denaturation , Serine Proteinase Inhibitors , Serpins
9.
Protein Expr Purif ; 22(3): 414-21, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11483003

ABSTRACT

C1 esterase inhibitor (C1INH) is an important regulator of the classical complement pathway. Hereditary deficiency of C1INH causes angioedema of the skin, gut, and respiratory tissues that may be fatal. C1INH replacement therapy may be lifesaving for patients with this disorder. The objective of this study was to evaluate the use of the baculovirus expression vector system for mass producing biologically active human recombinant (rC1INH). A recombinant baculovirus was constructed coding the human native (nC1INH) sequence under control of the polyhedrin promoter. Spodoptera frugiperda Sf-9 insect cells were infected with this recombinant baculovirus in a medium-scale (10-L) bioreactor to produce rC1INH with a specific activity of 45 U/mg. Purification of rC1INH from the culture harvested at 60 h postinfection yielded 5.9 microg rC1INH/mL supernatant of a 75-kDa product with a specific activity of 31,000 U/mg purified rC1INH compared to 71,000 U/mg purified nC1INH from human serum using the same procedure. This rC1INH was about 25 kDa smaller than nC1INH, suggesting that Sf-9 cells express underglycosylated rC1INH. Glycan analysis showed that both N-glycan and O-glycan chains were present in rC1INH. The N-glycan chains, released using PNGaseF and fluorescently labeled, were analyzed using exoglycosidase treatment and capillary electrophoresis. Their high-mannose structure was consistent with the known failure of the insect cell glycosylation pathway to afford the fully elaborated biantennary structures found on human native nC1INH.


Subject(s)
Complement C1 Inactivator Proteins/genetics , Animals , Baculoviridae/genetics , Complement C1 Inactivator Proteins/isolation & purification , Complement C1 Inactivator Proteins/metabolism , Genetic Vectors , Humans , Polysaccharides/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Spodoptera
10.
Protein Expr Purif ; 22(2): 349-58, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11437612

ABSTRACT

Human C1 inhibitor is a highly glycosylated serine protease inhibitor of the serpin family. The protein contains two disulfide bonds. In this study, an N-terminally truncated form of recombinant C1 inhibitor was overexpressed in Escherichia coli strains BL21(DE3) and AD494(DE3), the latter enabling the formation of disulfide bonds within the cytoplasm. With both strains, a major fraction of the recombinant protein produced appeared to be insoluble. However, the soluble fraction of lysates from strain AD494(DE3) inhibited the C1s target protease in functional assays. Recombinant C1 inhibitor produced in this strain also displayed the ability to complex with C1s in vitro. In contrast, lysates from strain BL21(DE3) displayed no C1 inhibitor activity. These data support the notion that glycosylation is not important, whereas disulfide bond formation appears to be essential for the production of an active recombinant C1 inhibitor. Thus, bacterial strains that permit the formation of disulfide bonds may represent a reliable system for the production of recombinant C1 inhibitor. However, a major obstacle to large-scale production will be to produce the protein in a soluble form. Attempts to increase the yield of soluble protein by coexpression of the GroEL/ES chaperonins resulted in an increase in solubility.


Subject(s)
Complement C1 Inactivator Proteins/genetics , Complement C1 Inactivator Proteins/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Amino Acid Sequence , Base Sequence , Chaperonin 10/biosynthesis , Chaperonin 10/genetics , Chaperonin 60/biosynthesis , Chaperonin 60/genetics , Complement C1 Inactivator Proteins/biosynthesis , Complement C1 Inactivator Proteins/isolation & purification , Complement Pathway, Classical/genetics , Escherichia coli/metabolism , Humans , Isopropyl Thiogalactoside/metabolism , Molecular Sequence Data , Peptide Fragments/biosynthesis , Peptide Fragments/isolation & purification , Plasmids/chemical synthesis , Plasmids/metabolism , Protein Structure, Tertiary/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Deletion , Serpins/biosynthesis , Serpins/genetics , Solubility , Temperature
11.
Thromb Haemost ; 85(2): 349-55, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11246559

ABSTRACT

In this study, the protein which stimulates proplatelet formation (PPF) of megakaryocytes was purified from normal human plasma using 7 steps procedures. Two different protease inhibitors were identified based on their amino acid sequences, i.e. antithrombin III (AT III) and C1 inhibitor. They were included in high density lipoprotein (HDL). HDL was necessary for AT III to be active in PPF in vitro. The biological effects of the AT III/HDL or thrombin-AT III (TAT)/HDL were studied in vitro. PPF of murine megakaryocytes was stimulated by negative control (BSA) (1.8 +/- 0.3%), AT III (2.0 +/- 0.4%), HDL (1.2 +/- 0.9%), AT III/HDL (14.8 +/- 2.1%) or TAT/HDL (23.3 +/- 3.5%), respectively. TAT/HDL also had a synergistic effect with the mpl ligand, judging by the acetylcholinesterase (AchE) expression of murine megakaryocytes (2.7 fold increase). In vivo subcutaneous administration of AT III alone or TAT for 3 days significantly stimulated thrombocytosis (136% and 144%, respectively, p<0.05) and AT III/HDL showed rapid and further stimulation (150%, p <0.01). These results and the previous studies indicate that megakaryocytopoiesis is regulated by the mpl ligand, while a protease/protease inhibitor complex such as TAT, which is involved in the coagulation cascade associated with platelet consumption, might be one of the regulators in platelet production.


Subject(s)
Antithrombin III/isolation & purification , Antithrombin III/pharmacology , Blood Platelets/drug effects , Complement C1 Inactivator Proteins/isolation & purification , Megakaryocytes/drug effects , Peptide Hydrolases/pharmacology , Amino Acid Sequence , Animals , Antithrombin III/drug effects , Antithrombin III/metabolism , Blood Platelets/cytology , Bone Marrow Cells/cytology , Cell Culture Techniques , Cell Line , Complement C1 Inactivator Proteins/drug effects , Complement C1 Inactivator Proteins/metabolism , Culture Media, Conditioned/chemistry , Hematologic Diseases/blood , Hematopoiesis/drug effects , Humans , Male , Megakaryocytes/cytology , Mice , Mice, Inbred Strains , Molecular Sequence Data
12.
Anal Biochem ; 248(2): 228-33, 1997 Jun 01.
Article in English | MEDLINE | ID: mdl-9177748

ABSTRACT

C1-inhibitor is a proteinase inhibitor in the serpin family. It is an important inhibitor of complement C1, plasma kallikrein, and factor XIIa, and as such is involved in regulating inflammatory pathways. Studies on the plasma-derived protein are hampered by the relative ease with which the protein converts to an inactive state on storage, under mild denaturing conditions, or by incubating in some unfavorable buffers. This inactivation is caused by formation of soluble polymers which can be visualized on native electrophoresis. In order to facilitate studies on both the plasma-derived protein and recombinant variants planned for the future, it was necessary to devise a method for the rapid reactivation of the polymers in high yield. It was found that nonionic detergents did not dissociate the polymers, but they were readily dissociated in 0.1% SDS. Treatment with 0.1% SDS followed by rapid removal of the SDS and refolding on an FPLC Superose 6 column allowed for recovery of about 15% of the protein in the active monomeric form. Eighty-five percent eluted as a range of higher order polymers. Using 8 M urea as the denaturant a 25% yield of active monomer was recovered. However, with 6 M guanidine hydrochloride as the denaturant, the yield of active monomer was almost 50%. The remaining material was not present as a range of polymeric species but was probably a dimer. Therefore this method is a useful technique to facilitate studies on C1-inhibitor. Moreover, the ability to produce monomer, dimer, and polymer forms of C1-inhibitor is useful for studies investigating the conformational changes which have occurred in the different forms.


Subject(s)
Complement C1 Inactivator Proteins/chemistry , Complement C1 Inactivator Proteins/isolation & purification , Chromatography, Gel , Complement C1 Inactivator Proteins/metabolism , Detergents , Dimerization , Guanidine , Guanidines , Humans , In Vitro Techniques , Polymers/chemistry , Polymers/isolation & purification , Protein Conformation , Protein Denaturation , Protein Folding , Urea
13.
Clin Exp Immunol ; 105(1): 191-7, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8697630

ABSTRACT

Clearance rates in the guinea pig were determined for intact guinea pig and human C1 inhibitor, the complexes of both inhibitors with human Cls, beta factor XIIa and kallikrein, and for each inhibitor cleaved at its reactive centre with trypsin. Intact human and guinea pig C1 inhibitor were cleared from the circulation more slowly (t1/2s of 9-7 h and 12.1 h and fractional catabolic rates (FCRs) of 0.09 and 0.117) than any of their cleaved or complexed forms. The reactive centre-cleaved inhibitors were cleared with half-lives of 6.75 h for humans and 10.1 h for the guinea pig. The complexes with target proteases were catabolized much more rapidly, with half-lives ranging from 3-08 h to 4.3 h. The complexes with kallikrein were cleared more slowly than those with Cls and beta factor XIIa. Complexes prepared with the guinea pig and human inhibitors were cleared at equivalent rates. The free inactivated proteases were cleared at rates similar to the equivalent complexes, except for kallikrein, which was cleared more rapidly than its complex. The fact that the complexes with different target proteases differed in their catabolism and that protease and complex catabolism were similar suggests that protease may play a direct role in clearance.


Subject(s)
Complement C1 Inactivator Proteins/pharmacokinetics , Endopeptidases/pharmacokinetics , Amino Acid Sequence , Animals , Binding Sites , Complement C1 Inactivator Proteins/chemistry , Complement C1 Inactivator Proteins/isolation & purification , Complement C1s/chemistry , Complement C1s/pharmacokinetics , Endopeptidases/chemistry , Factor XIIa/chemistry , Factor XIIa/pharmacokinetics , Guinea Pigs , Humans , Kallikreins/chemistry , Kallikreins/pharmacokinetics , Molecular Sequence Data
14.
Immunol Lett ; 42(3): 185-90, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7890318

ABSTRACT

Factor J (FJ) is a new inhibitor of the complement system. This work supports the fact that FJ is a cationic molecule (pI > or = 9.6 in native conditions, or pI = 8.1 in denaturing conditions) with a high sugar content (40%) that is able to interact with different lectins, suggesting a complex glycosylation. SDS impaired FJ migration in polyacrylamide gel electrophoresis. In Triton-acid-urea-polyacrylamide gel electrophoresis FJ migrated as a complex, dispersed molecule. In contrast, FJ after Smith degradation (dFJ) gave a single, smeared band of M(r) = 23.4 kDa in reducing SDS-PAGE. dFJ retained only 60% of the initial inhibitory activity of intact FJ. When digestions with different proteinases were performed, no modification of activity was observed. After beta-glucuronidase digestion, FJ lost 80% of its initial activity. Consequently, glycosylation plays an important role in the inhibitory activity of FJ.


Subject(s)
Complement C1 Inactivator Proteins/chemistry , Glycoproteins/chemistry , Carbohydrate Sequence , Chromatography, High Pressure Liquid , Complement C1 Inactivator Proteins/isolation & purification , Complement Hemolytic Activity Assay , Electrophoresis, Polyacrylamide Gel , Glycoproteins/isolation & purification , Humans , Isoelectric Point , Molecular Sequence Data
15.
Blood Coagul Fibrinolysis ; 5(4): 543-9, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7841310

ABSTRACT

A two-step chromatographic procedure has been developed to purify human C1-inhibitor from cryoprecipitate-poor plasma after removal of vitamin K-dependent proteins and antithrombin III. The procedure, which is fully compatible with modern plasma fractionation schemes, includes anion-exchange chromatography on DMAE-Fractogel EMD, viral inactivation by solvent-detergent treatment, adsorption on SO3-Fractogel EMD and viral removal by nanofiltration on 35- and 15-nm pore size membranes. Overall yields were about 45% and 58% for antigen and activity, respectively, providing 60-70 mg of highly purified inhibitor per litre of plasma. The purified inhibitor had a specific activity of 6.5 +/- 0.5 units/mg protein, representing a more than 400-fold increase in purity compared with plasma. C1-inhibitor purity with respect to total protein was greater than 80%. The main contaminant was complement component C3 which accounted for 4-10% of the total protein. Minor contaminants included low amounts of IgM, IgG, IgA, fibrinogen and albumin. Complement component C4 was undetectable. The purified inhibitor was stable throughout the purification process and for more than 24 h at room temperature after reconstitution of the freeze-dried material. Animal tests in rats and mice demonstrated that the C1-inhibitor concentrate was well tolerated at relatively high doses.


Subject(s)
Chromatography, Ion Exchange , Complement C1 Inactivator Proteins/isolation & purification , Animals , Complement C1 Inactivator Proteins/therapeutic use , Humans , Mice , Polysorbates , Rats , Solvents , Ultrafiltration , Viruses/drug effects
16.
Clin Biochem ; 27(3): 169-76, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7923756

ABSTRACT

Factor J (FJ) is a protein present in human serum, with inhibitory activity against C1. Here we describe the quantitation of FJ in human serum by means of an ELISA inhibition assay. We have purified FJ from the urine of a normal donor following a previously published method with slight modifications. Polyclonal anti-FJ antibodies have been raised in rabbits immunized with a single dose of purified antigen injected in multiple sites. IgG from polyclonal FJ antiserum, coupled to a solid matrix (Affi-Prep gel) was able to adsorb purified FJ antigenically and functionally. Furthermore, anti-FJ specifically retained serum components antigenically related with urine FJ. Taking into account this reactivity, we have developed an inhibition enzyme-linked immunosorbent assay (ELISA) useful for measuring FJ levels in normal human serum. This immunoassay involves preincubating polyclonal anti-FJ with different dilutions of normal human serum to quantitatively reduce the antibody available to bind to purified FJ-coated microtiter plates. Binding of remaining antibody to the microtiter plate is measured spectrophotometrically using peroxidase-conjugated secondary antibody. Quantitation is accomplished by comparison with a known quantity of purified FJ. Conditions for optimization of this quantitative assay have been assessed, including trials with different blocking agents, of which nonfat milk gave the best results. Preliminary experiments showed the existence of paradoxical effects, that is, high nonspecific binding at high serum dilutions. We have eliminated these effects by including high ionic strength (0.4 M NaCl) in the sample incubation solution. Sensitivity and reproducibility parameters have also been established. FJ levels have been measured for the first time in sera from 86 healthy donors.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Carrier Proteins/analysis , Complement C1 Inactivator Proteins/analysis , Complement Inactivator Proteins , Enzyme-Linked Immunosorbent Assay/methods , Glycoproteins/analysis , Animals , Antibodies, Monoclonal , Antibody Specificity , Carrier Proteins/immunology , Carrier Proteins/isolation & purification , Complement C1 Inactivator Proteins/immunology , Complement C1 Inactivator Proteins/isolation & purification , Glycoproteins/immunology , Glycoproteins/isolation & purification , Humans , Rabbits , Reference Values , Sensitivity and Specificity
17.
Behring Inst Mitt ; (93): 214-23, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8172570

ABSTRACT

The serum C1q inhibitor (C1q INH) is a chondroitin 4-sulfate proteoglycan which is composed of several polyanionic components ranging in size from 21-750 kDa. Although the activity of C1q INH has been described in terms of its ability to precipitate C1q and inhibit its hemolytic activity, not much is known about either the mechanisms of its action or its role in health and disease. This report provides evidence that a 30 kDa core protein component of the proteoglycan macromolecule contains most of the C1q inhibitory activity. This inhibitory activity occurs as a result of C1q INH binding to the C1q "heads" (gC1q) as well as to the collagen "tail" (cC1q). What may be more significant in terms of perpetuation of inflammatory processes is the ability of C1q INH to moderately activate the classical pathway leading to C2 and C4 consumption. The binding of C1q INH to C1q is enhanced at low ionic strength, but significant binding does occur under physiologic conditions which makes it likely for the inhibitor to participate in inflammatory processes especially in microenvironments of high inhibitor concentration. Such elevated concentration does occur in patients with active rheumatoid arthritis and systemic lupus erythematosus either as a result of unregulated proteoglycan synthesis or disturbances in connective tissue metabolism. Another important function of serum C1q INH is its ability to prolong the clotting time of plasma and fibrinogen solutions containing or lacking CaCl2. This potent anticoagulant activity is again displayed by the 30 kDa putative protein core which specifically binds to both the E and D domains of fibrinogen. However, the epitope(s) on the 30 kDa which binds to C1q appears to be distinct from that which binds to fibrinogen. The known presence of proteoglycans on the basement membranes and other sites may explain at least in part the presence of fibrinogen in atheromatous lesions. Furthermore, by binding to fibrinogen, soluble C1q INH-and C1q-C1q INH complexes may limit fibrin gelation in inflammatory and tissue repair microenvironments.


Subject(s)
Chondroitin Sulfate Proteoglycans/chemistry , Chondroitin Sulfate Proteoglycans/metabolism , Complement Activation , Complement C1 Inactivator Proteins/metabolism , Complement C1q/metabolism , Chondroitin Sulfate Proteoglycans/isolation & purification , Chromatography, Affinity , Complement C1 Inactivator Proteins/chemistry , Complement C1 Inactivator Proteins/isolation & purification , Humans , Kinetics , Molecular Weight
18.
Eur J Immunol ; 23(11): 2939-44, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8223871

ABSTRACT

Incubation of highly purified human C1 inhibitor with equally pure human leukocyte proteinase 3, resulted in a dose- and time-dependent inactivation of C1 inhibitor hemolytic activity. Furthermore, this inactivation was accompanied by proteinase 3-dependent cleavage of the C1 inhibitor into an 83,000 molecular weight fragment. The formation of the 83,000 molecular weight fragment followed a time course which was similar to that observed for the inactivation of hemolytic activity. Within 120 minutes more than 90% of the hemolytic activity was lost. This inactivation of C1 inhibitor appeared to be selective as purified human C1q was not degraded in a similar time period. Moreover, when 100 micrograms IgG, isolated from each of 21 Wegener's granulomatosis patients with cytoplasmic anti-nuclear antibodies immunofluorescent titers to proteinase 3 greater then 1:64, was incubated with 3 milliunits of proteinase 3, little to no cleavage of C1 inhibitor was observed. In contrast, 100 micrograms of IgG isolated from 14 normal donors was ineffective in affording protection to C1 inhibitor upon incubation with proteinase 3. Our results suggest that neutrophil infiltration and activation could lead to local complement consumption at the tissue sites.


Subject(s)
Complement C1 Inactivator Proteins/metabolism , Serine Endopeptidases/pharmacology , Antibodies, Antineutrophil Cytoplasmic , Autoantibodies/pharmacology , Complement C1 Inactivator Proteins/isolation & purification , Granulomatosis with Polyangiitis/immunology , Humans , Immunoglobulin G/pharmacology , In Vitro Techniques , Kinetics , Molecular Weight , Myeloblastin , Serine Endopeptidases/immunology
19.
J Biol Chem ; 268(24): 18088-94, 1993 Aug 25.
Article in English | MEDLINE | ID: mdl-8349686

ABSTRACT

C1-inhibitor(Mo), a dysfunctional C1-inhibitor molecule produced in two kindred with type II hereditary angioedema, has a mutation at the P10 position (Ala436 to Thr). Like most serpins with hinge region mutations (P14, P12, P10), C1-inhibitor(Mo) loses its inhibitory activity. However, unlike the other hinge region mutations, this mutant is not converted to a substrate. As shown by nondenaturing gel electrophoresis, gel filtration, sucrose density gradient ultracentrifugation, and electron microscopy, C1-inhibitor(Mo) exists in both monomeric and multimeric forms. Polymerization probably results from reactive center loop insertion into the A sheet of an adjacent molecule. Native C1-inhibitor(Mo) was shown to have a thermal stability profile intermediate to those of intact and of cleaved normal C1-inhibitor. Native C1-inhibitor(Mo) did not bind to monoclonal antibody KII, which binds only to reactive center-cleaved normal C1-inhibitor. It did, however, react with monoclonal antibody KOK12, which recognizes complexed or cleaved C1-inhibitor but not intact normal C1-inhibitor. Native C1-inhibitor(Mo), therefore, exists in a conformation similar to the complexed form of normal C1-inhibitor.


Subject(s)
Alanine , Complement C1 Inactivator Proteins/chemistry , Complement C1 Inactivator Proteins/genetics , Point Mutation , Amino Acid Sequence , Animals , Cell Line , Centrifugation, Density Gradient , Chromatography, Gel , Cloning, Molecular , Complement C1 Inactivator Proteins/isolation & purification , Drug Stability , Electrophoresis, Polyacrylamide Gel , Humans , Microscopy, Electron , Protein Denaturation , Thermodynamics , Threonine , Transfection
20.
J Clin Invest ; 91(3): 1258-62, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8450054

ABSTRACT

To ascertain the mechanism for decreased synthesis of C1 inhibitor (C1 INH) in certain patients with the autosomal dominant disorder hereditary angioneurotic edema, we studied expression of C1 INH in fibroblasts in which the mutant and wild type mRNA and protein could be distinguished because of deletion of exon 7 (delta Ex7). In the HANE delta Ex7 cells, the amount of wild type mRNA (2.1 kb) was expressed at 52 +/- 2% (n = 5) of normal, whereas the mutant mRNA was 17 +/- 1% (n = 5) of normal. Rates of synthesis of both wild type and mutant proteins (11 +/- 3 and 3 +/- 1% of normal, respectively) were lower than predicted from the mRNA levels. There was no evidence of increased C1 INH protein catabolism. These data indicate that there are multiple levels of control of C1 INH synthesis in type I hereditary angioneurotic edema. Pretranslational regulation results in < 50% of the mutant truncated 1.9-kb mRNA. In addition, translational regulation results in decreased synthesis of both wild type and mutatn C1 INH proteins. These data suggest a transinhibition of wild type C1 INH translation by mutant mRNA and/or protein.


Subject(s)
Angioedema/metabolism , Complement C1 Inactivator Proteins/biosynthesis , Adult , Angioedema/genetics , Cell Line , Cells, Cultured , Complement C1 Inactivator Proteins/genetics , Complement C1 Inactivator Proteins/isolation & purification , Exons , Fibroblasts/metabolism , Humans , Kinetics , Molecular Weight , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reference Values , Sequence Deletion , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...