Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Osteoarthritis Cartilage ; 17(4): 547-55, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18930415

ABSTRACT

UNLABELLED: Insulin-like growth factor-I (IGF-I) and IGF binding proteins (IGFBPs) are trophic factors for cartilage and have been shown to be chondroprotective in animal models of osteoarthritis (OA). IGFBP-5 is degraded in joint fluid and inhibition of IGFBP-5 degradation has been shown to enhance the trophic effects of IGF-I. OBJECTIVE: To determine the identity of IGFBP-5 protease activity in human OA joint fluid. METHOD: OA joint fluid was purified and the purified material was analyzed by IGFBP-5 zymography. RESULTS: Both crude joint fluid and purified material contained a single band of proteolytic activity that cleaved IGFBP-5. Immunoblotting of joint fluid for complement 1s (C1s) showed a band that had the same Mr estimate, e.g., 88 kDa. In gel tryptic digestion and subsequent peptide analysis by LC-MS/MS showed that the band contained human C1s. A panel of protease inhibitors was tested for their ability to inhibit IGFBP-5 cleavage by the purified protease. Three serine protease inhibitors, FUT175 and CP-143217 and CB-349547 had IC50's between 1 and 6 microM. Two other serine protease inhibitors had intermediate activity (e.g., IC50's 20-40 microM) and MMP inhibitors had no detectible activity at concentrations up to 300 microM. CONCLUSION: Human OA fluid contains a serine protease that cleaves IGFBP-5. Zymography, immunoblotting and LC-MS/MS analysis indicate that C1s is the protease that accounts for this activity.


Subject(s)
Complement C1s/physiology , Insulin-Like Growth Factor Binding Protein 5/metabolism , Osteoarthritis, Knee/metabolism , Serine Proteases/physiology , Synovial Fluid/metabolism , Complement C1s/antagonists & inhibitors , Complement C1s/metabolism , Humans , Osteoarthritis, Knee/enzymology , Peptide Fragments/metabolism , Serine Proteases/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology
3.
J Immunol ; 175(7): 4536-42, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16177097

ABSTRACT

C1s is the modular serine protease responsible for cleavage of C4 and C2, the protein substrates of the first component of C (C1). Its catalytic domain comprises two complement control protein (CCP) modules connected by a four-residue linker Gln340-Pro-Val-Asp343 and a serine protease domain. To assess the functional role of the linker, a series of mutations were performed at positions 340-343 of human C1s, and the resulting mutants were produced using a baculovirus-mediated expression system and characterized functionally. All mutants were secreted in a proenzyme form and had a mass of 77,203-77,716 Da comparable to that of wild-type C1s, except Q340E, which had a mass of 82,008 Da, due to overglycosylation at Asn391. None of the mutations significantly altered C1s ability to assemble with C1r and C1q within C1. Whereas the other mutations had no effect on C1s activation, the Q340E mutant was totally resistant to C1r-mediated activation, both in the fluid phase and within the C1 complex. Once activated, all mutants cleaved C2 with an efficiency comparable to that of wild-type C1s. In contrast, most of the mutations resulted in a decreased C4-cleaving activity, with particularly pronounced inhibitory effects for point mutants Q340K, P341I, V342K, and D343N. Comparable effects were observed when the C4-cleaving activity of the mutants was measured inside C1. Thus, flexibility of the C1s CCP1-CCP2 linker plays no significant role in C1 assembly or C1s activation by C1r inside C1 but plays a critical role in C4 cleavage by adjusting positioning of this substrate for optimal cleavage by the C1s active site.


Subject(s)
Complement C1s/physiology , Amino Acid Motifs , Amino Acid Substitution/genetics , Animals , Asparagine/metabolism , Baculoviridae , Complement C1s/genetics , Complement C2/metabolism , Complement C4/metabolism , Genetic Vectors , Glycosylation , Humans , Kinetics , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Spodoptera
4.
J Immunol ; 173(1): 349-59, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15210793

ABSTRACT

There is growing evidence that certain components of complement systems in lower vertebrates are promiscuous in their modes of activation through the classical or alternative pathways. To better understand the evolution of the classical pathway, we have evaluated the degree of functional diversification of key components of the classical and alternative pathways in rainbow trout, an evolutionarily relevant teleost species. Trout C4 was purified in two distinct forms (C4-1 and C4-2), both exhibiting the presence of a thioester bond at the cDNA and protein levels. C4-1 and C4-2 bound in a similar manner to trout IgM-sensitized sheep erythrocytes in the presence of Ca(2+)/Mg(2+), and both C4 molecules equally restored the classical pathway-mediated hemolytic activity of serum depleted of C3 and C4. Reconstitution of activity was dependent on the presence of both C3-1 and C4-1/C4-2 and on the presence of IgM bound to the sheep erythrocytes. A C1s-like molecule was shown to cleave specifically purified C4-1 and C4-2 into C4b, while failing to cleave trout C3 molecules. The C1s preparation was unable to cleave trout factor B/C2 when added in the presence of C3b or C4b molecules. Our results show a striking conservation of the mode of activation of the classical pathway. We also show that functional interchange between components of the classical and alternative pathway in teleosts is more restricted than was anticipated. These data suggest that functional diversification between the two pathways must have occurred shortly after the gene duplication that gave rise to the earliest classical pathway molecules.


Subject(s)
Complement C1s/chemistry , Complement C4/chemistry , Complement Pathway, Alternative , Complement Pathway, Classical , Oncorhynchus mykiss/immunology , Amino Acid Sequence , Animals , Base Sequence , Biological Evolution , Complement C1s/physiology , Complement C4/genetics , Complement C4/physiology , DNA, Complementary/isolation & purification , Glycosylation , Hemolysis , Methylamines/metabolism , Molecular Sequence Data
6.
J Immunol ; 170(9): 4701-7, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12707349

ABSTRACT

Mannose-binding lectin-associated serine proteases (MASPs) are involved in complement activation through the lectin pathway. To elucidate the phylogenetic origin of MASP and a primordial complement system, we cloned two MASP cDNAs from amphioxus (Branchiostoma belcheri) of the cephalochordates, considered to be the closest relative of vertebrates. The two sequences, orthologues of mammalian MASP-1 and MASP-3, were produced by alternative processing of RNA from a single gene consisting of a common H chain-encoding region and two L chain-encoding regions, a structure which is similar to that of the human MASP1/3 gene. We also isolated two MASP genes from the ascidian Halocynthia roretzi (urochordates) and found that each of them consists simply of an H chain-encoding region and a single L chain-encoding region. The difference in structure between the ascidian MASP genes and the amphioxus/mammalian MASP genes suggests that a prototype gene was converted to the MASP1/3-type gene possessing two L chain-encoding regions at an early stage of evolution before the divergence of amphioxus. This conclusion is supported by the presence of MASP-1 and MASP-3 homologues in almost all vertebrates, as demonstrated by the cloning of novel cDNA sequences representing lamprey (cyclostomes) MASP-1 and Xenopus MASP-3. The ancient origin of MASP-1 and MASP-3 suggests that they have crucial functions common to all species which emerged after cephalochordates.


Subject(s)
Chordata, Nonvertebrate/immunology , Complement Activation , Evolution, Molecular , Lectins/metabolism , Mannose/metabolism , Serine Endopeptidases/isolation & purification , Serine Endopeptidases/physiology , Amino Acid Sequence , Animals , Chordata, Nonvertebrate/genetics , Cloning, Molecular , Complement Activation/genetics , Complement C1r/chemistry , Complement C1r/genetics , Complement C1r/physiology , Complement C1s/chemistry , Complement C1s/genetics , Complement C1s/physiology , Consensus Sequence , DNA, Complementary/isolation & purification , Genes , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/isolation & purification , Isoenzymes/physiology , Lampreys/genetics , Lampreys/immunology , Mannose-Binding Protein-Associated Serine Proteases , Molecular Sequence Data , Multigene Family/genetics , Multigene Family/immunology , Phylogeny , Sequence Homology, Amino Acid , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Urochordata/genetics , Urochordata/immunology , Xenopus/genetics , Xenopus/immunology
7.
Immunobiology ; 205(4-5): 383-94, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12396001

ABSTRACT

C1s is the modular serine protease, which executes the catalytic function of the C1 complex: the cleavage of C4 and C2. Like other complement serine proteases C1s has restricted substrate specificity and it is engaged into specific interactions with other subcomponents of the complement system. There has been a rapid progress in determining the 3D structure of complement serine proteases and in revealing the role of the individual domains in the protein-protein interaction properties. In this review we summarize recent findings on the structure of C1s, and on the mechanism of action of this protease. The results obtained by genetic engineering, physico-chemical and functional studies are reviewed. The physiological relevance of the proteolytic action of C1s and its possible implications in health and disease will also be discussed.


Subject(s)
Complement C1s/chemistry , Complement C1s/physiology , Complement Pathway, Classical/physiology , Structure-Activity Relationship , Animals , Complement C1s/genetics , Enzyme Activation , Humans , Protein Structure, Secondary
8.
Immunobiology ; 205(4-5): 365-82, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12396000

ABSTRACT

C1r, the enzyme responsible for intrinsic activation of the C1 complex of complement, is a modular serine protease featuring an overall structural organization homologous to those of C1s and the mannan-binding lectin-associated serine proteases (MASPs). This review will initially summarize current information on the structure and function of C1r, with particular emphasis on the three-dimensional structure of its catalytic domain, which provides new insights into the activation mechanism of C1. The second part of this review will focus on recent discoveries dealing with a truncated, C1r-related protein, and the occurrence in the mouse of two isoforms, C1rA and C1rB, exhibiting tissue-specific expression patterns.


Subject(s)
Complement C1r/chemistry , Complement C1r/genetics , Complement C1r/physiology , Complement Pathway, Classical/physiology , Animals , Complement C1s/chemistry , Complement C1s/physiology , Enzyme Activation , Humans , Mice , Protein Isoforms , Protein Structure, Secondary , Structure-Activity Relationship
9.
Microvasc Res ; 62(3): 284-92, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11678631

ABSTRACT

Recent observations provide evidence that complement is implicated as an important factor in the pathophysiology of ischemia/reperfusion injury (IRI). Here, we assessed the effects of complement inhibition on hepatic microcirculation by in vivo microscopy (IVM) using a rat model of warm hepatic ischemia clamping the left pedicle for 70 min. Ten animals received the physiological complement regulator soluble complement receptor type 1 (sCR1) intravenously 1 min prior to reperfusion. Controls were given an equal amount of Ringer's solution (n = 10). Microvascular perfusion and leukocyte adhesion were studied 30 to 100 min after reperfusion by IVM. Microvascular perfusion in hepatic sinusoids was significantly improved in the sCR1 group (80.6 +/- 0.6% of all observed sinusoids were perfused [sCR1] vs 67.3 +/- 1.2% [controls]). The number of adherent leukocytes was reduced in sinusoids (49.9 +/- 3.4 [sCR1] vs 312.3 +/- 14.2 in controls [adherent leukocytes per square millimeter of liver surface]; P < 0.001) as well as in postsinusoidal venules after sCR1 treatment (230.9 +/- 21.7 [sCR1] vs 1906.5 +/- 93.5 [controls] [adherent leukocytes per square millimeter of endothelial surface]; P < 0.001). Reflecting reduced hepatocyte injury, liver transaminases were decreased significantly upon sCR1 treatment compared to controls. Our results provide further evidence that complement plays a decisive role in warm hepatic IRI. Therefore, we conclude that complement inhibition by sCR1 is effective as a therapeutical approach to reduce microcirculatory disorders after reperfusion following warm organ ischemia.


Subject(s)
Complement C1 Inactivator Proteins/pharmacology , Complement C1s/physiology , Ischemia/physiopathology , Liver Circulation/physiology , Microcirculation/drug effects , Receptors, Complement/physiology , Reperfusion Injury/physiopathology , Animals , Cell Adhesion/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Hemodynamics , Injections, Intravenous , Ischemia/pathology , Laser-Doppler Flowmetry/instrumentation , Leukocytes/physiology , Liver Circulation/drug effects , Male , Microcirculation/physiopathology , Microscopy, Fluorescence , Rats , Rats, Wistar , Receptors, Complement/administration & dosage , Temperature , Time Factors , Venules/physiopathology , Video Recording
10.
J Immunol ; 166(12): 7612-6, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11390518

ABSTRACT

We have investigated the molecular basis of selective and complete C1s deficiency in 2-year-old girl with complex autoimmune diseases including lupus-like syndrome, Hashimoto's thyroiditis, and autoimmune hepatitis. This patient's complement profile was characterized by the absence of CH50 activity, C1 functional activity <10%, and undetectable levels of C1s Ag associated with normal levels of C1r and C1q Ags. Exon-specific amplification of genomic DNA by PCR followed by direct sequence analysis revealed a homozygous nonsense mutation in the C1s gene exon XII at codon 534, caused by a nucleotide substitution from C (CGA for arginine) to T (TGA for stop codon). Both parents were heterozygous for this mutation. We used the new restriction site for endonuclease Fok-1 created by the mutation to detect this mutation in the genomic DNA of seven healthy family members. Four additional heterozygotes for the mutation were identified in two generations. Our data characterize for the first time the genetic defect of a selective and complete C1s deficiency in a Caucasian patient.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Complement C1s/deficiency , Complement C1s/genetics , Age of Onset , Autoimmune Diseases/enzymology , Base Sequence , Blotting, Western , Child, Preschool , Codon, Nonsense/immunology , Complement C1s/isolation & purification , Complement C1s/physiology , Complement Hemolytic Activity Assay , Complement Pathway, Alternative/genetics , Complement Pathway, Classical/genetics , Exons/genetics , Female , Homozygote , Humans , Male , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Serine Endopeptidases/genetics
11.
J Biol Chem ; 275(48): 37638-44, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-10982804

ABSTRACT

Cultured fibroblasts secrete an 88-kDa serine protease that cleaves insulin-like growth factor binding protein-5 (IGFBP-5). Because IGFBP-5 has been shown to regulate IGF-I actions, understanding the chemical identity and regulation of this protease is important for understanding how IGF-I stimulates anabolic functions. The protease was purified from human fibroblast-conditioned medium by hydrophobic interaction, lectin affinity, and heparin Sepharose affinity chromatography followed by SDS-polyacrylamide gel electrophoresis. An 88-kDa band was excised and digested with lysyl-endopeptidase. Sequencing of the high pressure liquid chromatography-purified peptides yielded the complement components C1r and C1s. To confirm that C1r/C1s accounted for the proteolytic activity in the medium, immunoaffinity chromatography was performed. Most of the protease activity adhered to the column, and the eluant was fully active in cleaving IGFBP-5. SDS-polyacrylamide gel electrophoresis with silver staining showed two bands, and IGFBP-5 zymography showed a single 88-kDa band. Amino acid sequencing confirmed that the 88-kDa band contained only C1r and C1s. C1r in the fibroblast medium underwent autoactivation, and the activated form cleaved C1s. C1s purified from the conditioned medium cleaved C(4), a naturally occurring substrate. The purified protease cleaved IGFBP-5 but had no activity against IGFBP-1 through -4. C1 inhibitor, a protein known to inhibit activated C1s, was shown to inhibit the cleavage of IGFBP-5 by the protease in the conditioned medium. In summary, human fibroblasts secrete C1r and C1s that actively cleave IGFBP-5. The findings define a mechanism for cleaving IGFBP-5 in the culture medium, thus allowing release of IGF-I to cell surface receptors.


Subject(s)
Complement C1s/physiology , Insulin-Like Growth Factor Binding Protein 5/metabolism , Amino Acid Sequence , Cells, Cultured , Complement C1s/chemistry , Culture Media , Fibroblasts/metabolism , Humans , Hydrolysis , Molecular Sequence Data , Peptide Fragments/chemistry
12.
Inflamm Res ; 49(1): 42-6, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10778920

ABSTRACT

OBJECTIVE: To obtain a synthetic anti-complement inhibitor which has stronger activity than FUT-175 (nafamostat mesilate), as a synthetic ester derivative containing amidino and guanidino groups. METHODS: We synthesized several modified compounds of FUT-175. The anti-complement activities were measured using synthetic substrates and complement-mediated hemolysis in vitro. The anti-complement activity in vivo was evaluated via Forssman systemic shock in guinea pigs. RESULTS: FUT-175 inhibited C1r and C1s with IC50s of 1.7x10(-6) and 3.2x10(-7) M, respectively. Inhibitory activities were decreased by substitution of the amidino group with a hydrogen atom (compound 2), but not the guanidino group with a hydrogen atom (compound 3). Compound 6, in which the benzene ring of compound 3 was substituted with a furan ring, inhibited C1r and the complement-mediated hemolysis in high-diluted serum with higher potency than FUT-175. The inhibitory activity of compound 6 in hemolysis was weakened in low diluted serum. Compound 7 had a guanidino group inserted into compound 6; however, Compound 7 strongly inhibited hemolysis even in low-diluted serum, and suppressed Forssman systemic shock more potently than both FUT-175 and compound 6. CONCLUSIONS: These data suggest that the 2-furylcarboxylic acid derivatives have a strong potential for inhibiting the activities of the complement, and the guanidino group was required to retain high inhibitory activities in vivo, and compound 7 is a hopeful anti-complement agent.


Subject(s)
Complement C1 Inactivator Proteins/chemical synthesis , Complement C1 Inactivator Proteins/pharmacology , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/pharmacology , Trypsin Inhibitors/chemical synthesis , Trypsin Inhibitors/pharmacology , Animals , Benzamidines , Complement C1r/physiology , Complement C1s/physiology , Guanidines/chemistry , Guanidines/pharmacology , Guinea Pigs , Hemolysis , Male , Molecular Structure , Shock/drug therapy , Structure-Activity Relationship
13.
Immunopharmacology ; 42(1-3): 3-13, 1999 May.
Article in English | MEDLINE | ID: mdl-10408360

ABSTRACT

C1r and C1s, the proteases responsible for activation and proteolytic activity of the C1 complex of complement, share similar overall structural organizations featuring five nonenzymic protein modules (two CUB modules surrounding a single EGF module, and a pair of CCP modules) followed by a serine protease domain. Besides highly specific proteolytic activities, both proteases exhibit interaction properties associated with their N-terminal regions. These properties include the ability to bind Ca2+ ions with high affinity, to associate with each other within a Ca2+-dependent C1s-C1r-C1r-C1s tetramer, and to interact with C1q upon C1 assembly. Precise functional mapping of these regions has been achieved recently, allowing identification of the domains responsible for these interactions, and providing a comprehensive picture of their structure and function. The objective of this article is to provide a detailed and up-to-date overview of the information available on these domains, which are keystones of the assembly of C1, and appear to play an essential role at the interface between the recognition function of C1 and its proteolytic activity.


Subject(s)
Complement C1r/physiology , Complement C1s/physiology , Complement C1r/chemistry , Complement C1s/chemistry , Protein Conformation , Structure-Activity Relationship
14.
Immunobiology ; 199(2): 303-16, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9777414

ABSTRACT

C1r and C1s, the enzymes responsible for the activation and proteolytic activity of the C1 complex of complement, are modular serine proteases featuring similar overall structural organizations, yet expressing very distinct functional properties within C1. This review will initially summarize available information on the structure and function of the protein modules and serine protease domains of C1r and C1s. It will then focus on the regions of both proteases involved in: (i) assembly of C1s-C1r-C1r-C1s, the Ca(2+)-dependent tetrameric catalytic subunit of C1; (ii) expression of C1 catalytic activities. Particular emphasis will be aid on recent structural and functional studies that provide new insights into the complex mechanisms involved in the assembly, activation, and proteolytic activity of C1.


Subject(s)
Complement C1r/physiology , Complement C1s/physiology , Calcium/physiology , Catalysis , Complement C1r/chemistry , Complement C1s/chemistry , Enzyme Activation , Humans , Macromolecular Substances , Models, Molecular , Protein Conformation , Structure-Activity Relationship
15.
Immunobiology ; 199(2): 317-26, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9777415

ABSTRACT

Our protein engineering studies on human C1r and C1s revealed important characteristics of the individual domains of these multidomain serine-proteases, and supplied evidence about the cooperation of the domains to create binding sites, and to control the activation process. We expressed the recombinant subcomponents in the baculovirus-insect cell system and checked the biological activity. Deletions and point mutants of C1r were constructed and C1r-C1s chimeras were also produced. Our deletion mutants demonstrated that the N-terminal CUB domain and the EGF-like domain of C1r together are responsible for the calcium dependent C1r-C1s interaction. It seems very likely that these two modules form the calcium-binding site of the C1r alpha-fragment and participate in the tetramer formation. The deletion mutants also demonstrated that the N-terminal region of the C1r molecule contains essential elements involved in the control of activation of the serine-protease module. The substrate specificity of the serine-protease is also determined by the five N-terminal noncatalytic domain of C1r/C1s chimera, which contains the catalytic domain of C1s preceded by the N-terminal region of C1r, could replace the C1r in the hemolytically active C1 complex. The C1s/C1r chimera, in which the alpha-fragment of the C1r was replaced for that of the C1s exibits both C1r- and C1s-like characteristics. We stabilized the zymogen form of human C1r by mutating the Arg(463)-Ile(464) bond. Using our stable zymogen C1r we showed that one active C1r in the C1 complex is sufficient for the full activity of the entire complex. Further experiment with this mutant could provide us with important information about the structure of the C1 complex.


Subject(s)
Complement C1r/physiology , Complement C1s/physiology , Amino Acid Substitution , Animals , Binding Sites , Calcium/metabolism , Catalysis , Complement C1r/chemistry , Complement C1r/genetics , Complement C1s/chemistry , Complement C1s/genetics , Enzyme Activation , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Genetic Vectors/genetics , Humans , Models, Molecular , Moths/cytology , Mutagenesis, Site-Directed , Nucleopolyhedroviruses/genetics , Point Mutation , Protein Conformation , Protein Engineering , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Substrate Specificity
16.
J Immunol ; 155(5): 2642-51, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7650393

ABSTRACT

Previous studies showed that simply inserting or substituting a few amino acid residues immediately downstream of the proteolytic activation site in C component C3 renders that site susceptible to the C4-specific C1s protease. This report describes the results of extending those studies to the closely related component C5. We found that small changes, similar to those that made C3 susceptible to C1s, were insufficient to render C5 C1s-sensitive; and neither more extensive substitution downstream of the cleavage site with a 14 residue long segment from C4, nor upstream substitution with an 8 residue long C4 segment gave C1s cleavage. However, substitution of both the upstream and downstream segments gave a hybrid C5 protein, designated ASC4, which was cleaved by C1s. The protease sensitivity of ASC4 was curious, however, in that C1s was more active against the secreted extracellular biosynthetic precursor, pro-ASC4(E) than mature ASC4, whereas a C5-specific convertase cleaved the mature protein but not the precursor. In contrast, both mature and precursor forms of wild-type C5 were cleaved by the C5 convertase, but neither of course is recognized by C1s. These results demonstrate that a mutant C5 molecule can be constructed that is cleaved at the activation site by both C1s and C5 convertase. This suggests that the structures necessary for specific recognition by the two proteases have little or no overlap and that recognition by C5 convertase involves residues that are distant from the activation site itself.


Subject(s)
Complement C1s/physiology , Complement C5/genetics , Complement C5/metabolism , Amino Acid Sequence , Animals , Complement C1 Inactivator Proteins , Complement C3/metabolism , Complement C4/metabolism , Complement Factor B/physiology , Genetic Engineering , Mice , Molecular Sequence Data , Mutation , Rabbits
17.
Eur J Immunol ; 25(8): 2206-10, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7664783

ABSTRACT

A membrane-associated receptor for the C1q subcomponent of complement is widely distributed among different cell types. While a number of possible physiological functions of the C1q receptor (C1qR) on different cell types have been described, the way in which C1qR regulates complement activity remains unclear. This report describes the mechanism by which C1qR regulates activation of the first component of complement, C1. Using purified components of complement, we were able to show that membrane-associated C1qR as well as detergent-solubilized C1qR, purified from polymorphonuclear leukocytes, human umbilical vein endothelial cells or an endothelial cell line, EA.hy 926, are able to inhibit complement-mediated lysis of C1q-sensitized erythrocytes. Using hemolytic assays, we were able to demonstrate that C1qR prevents the association of C1q with C1r and C1s to form macromolecular C1. In addition, incubation of C1qR with the collagen-like stalks, but not with the globular heads of C1q, inhibits the effect of C1qR. This demonstrates that C1qR exerts its complement inhibitory effect by binding to the collagen-like stalk of C1q. No complement regulatory effect of C1qR was observed on preformed macromolecular C1. These data suggest that besides such-well-known complement regulatory molecules as CD55 (DAF), CD46 (MCP), CD35 (CR1) and CD59 (HRF), C1qR too is able to regulate complement activity.


Subject(s)
Complement C1 Inactivator Proteins/physiology , Hyaluronan Receptors , Membrane Glycoproteins , Receptors, Complement/physiology , Carrier Proteins , Complement C1q/antagonists & inhibitors , Complement C1r/antagonists & inhibitors , Complement C1s/physiology , Endothelium, Vascular/physiology , Humans , Mitochondrial Proteins , Neutrophils/physiology
19.
J Clin Invest ; 82(2): 700-5, 1988 Aug.
Article in English | MEDLINE | ID: mdl-2457036

ABSTRACT

The reactive center of C1-inhibitor, a plasma protease inhibitor that belongs to the serpin superfamily, is located on a peptide loop which is highly susceptible to proteolytic cleavage. With plasma kallikrein, C1s and beta-Factor XIIa, this cleavage occurs at the reactive site residue P1 (Arg444); with neutrophil elastase, it takes place near P1, probably at residue P3 (Val442). After these cleavages, C1-inhibitor is inactivated and its conformation is modified. Moreover, in vivo, cleaved C1-inhibitor is removed from the blood stream more rapidly than the intact serpin, which suggests that proteolysis unmasks sites responsible for cellular recognition and the uptake of the cleaved inhibitor. In the study reported here, we show, using an MAb, that an identical neoepitope is created on C1-inhibitor after the cleavage of its exposed loop by plasma kallikrein, C1s, beta-Factor XIIa, and by neutrophil elastase.


Subject(s)
Complement Activating Enzymes/metabolism , Complement C1 Inactivator Proteins/metabolism , Complement C1s/metabolism , Epitopes/isolation & purification , Factor XII/metabolism , Kallikreins/blood , Neutrophils/enzymology , Pancreatic Elastase/metabolism , Serine Endopeptidases/metabolism , Animals , Antibodies, Monoclonal , Complement C1 Inactivator Proteins/physiology , Complement C1s/physiology , Enzyme Activation , Factor XII/physiology , Factor XIIa , Hot Temperature , Humans , Immunoassay , Kallikreins/physiology , Mice , Mice, Inbred BALB C , Molecular Weight , Pancreatic Elastase/physiology , Peptide Fragments/metabolism , Peptide Fragments/physiology , Protein Conformation , Protein Denaturation , Serine Endopeptidases/physiology
20.
Nouv Presse Med ; 8(20): 1657-60, 1979 May 05.
Article in French | MEDLINE | ID: mdl-112575

ABSTRACT

Titrations of total complement (CH50) and of the different components of the classical pathway of the complement system in patients with supposed consumption hypocomplementemia, show that the complement depression involves in an order of decreasing severity hemolytic C4, hemolytic C2, total complement, hemolytic C1, protein C4 and protein C3. These results as well as correlative studies between these different parameters suggest that C4 is the most specific target of activited C1 esterase (C1s). They stress the interest of hemolytic titrations as well as the strong limitations of protein titrations.


Subject(s)
Blood Protein Disorders/diagnosis , Complement Activation , Complement Fixation Tests , Complement Pathway, Classical , Complement System Proteins/analysis , Adolescent , Adult , Aged , Child , Complement C1/analysis , Complement C1s/physiology , Complement C2/analysis , Complement C3/analysis , Complement C4/analysis , Complement Fixation Tests/methods , Complement System Proteins/metabolism , Female , Humans , Immune System Diseases/diagnosis , Immunodiffusion , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...