Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Sci Rep ; 11(1): 22511, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795372

ABSTRACT

Atherosclerotic cardiovascular disease (ACVD) is a lipid-driven inflammatory disease and one of the leading causes of death worldwide. Lipid deposits in the arterial wall lead to the formation of plaques that involve lipid oxidation, cellular necrosis, and complement activation, resulting in inflammation and thrombosis. The present study found that homozygous deletion of the CFHR1 gene, which encodes the plasma complement protein factor H-related protein 1 (FHR-1), was protective in two cohorts of patients with ACVD, suggesting that FHR-1 accelerates inflammation and exacerbates the disease. To test this hypothesis, FHR-1 was isolated from human plasma and was found to circulate on extracellular vesicles and to be deposited in atherosclerotic plaques. Surface-bound FHR-1 induced the expression of pro-inflammatory cytokines and tissue factor in both monocytes and neutrophils. Notably, plasma concentrations of FHR-1, but not of factor H, were significantly (p < 0.001) elevated in patients with ACVD, and correlated with the expression of the inflammation markers C-reactive protein, apolipoprotein serum amyloid protein A, and neopterin. FHR-1 expression also significantly correlated with plasma concentrations of low-density lipoprotein (LDL) (p < 0.0001) but not high-density lipoprotein (HDL). Taken together, these findings suggest that FHR-1 is associated with ACVD.


Subject(s)
Atherosclerosis/metabolism , Cardiovascular Diseases/metabolism , Complement C3b Inactivator Proteins/physiology , Gene Expression Regulation , Aged , Cardiology , Chromosome Deletion , Complement Activation , Complement C3b Inactivator Proteins/biosynthesis , Complement C3b Inactivator Proteins/genetics , Female , Gene Expression Profiling , Homozygote , Humans , Inflammation , Lipids/chemistry , Male , Middle Aged , Necrosis , Oxygen/chemistry , Sequence Deletion
2.
C R Biol ; 337(3): 178-84, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24702844

ABSTRACT

Age-related macular degeneration (AMD) is the major cause of legal blindness in the industrialized world. Polymorphisms and recently discovered rare mutations of the Complement Factor H gene have been shown to be strongly associated with AMD. The deletion of CFH-related proteins 1 and 3, proteins that share homologous regions with CFH, is found in protective haplotypes. The following is a critical review of the current state of knowledge of the implication of CFH and CFH-related proteins 1 and 3 in AMD.


Subject(s)
Complement Factor H/genetics , Macular Degeneration/drug therapy , Aging/physiology , Blood Proteins/genetics , Blood Proteins/physiology , Complement C3b Inactivator Proteins/genetics , Complement C3b Inactivator Proteins/physiology , Humans , Macular Degeneration/genetics
3.
Blood ; 114(12): 2439-47, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19528535

ABSTRACT

Homozygous deletion of a 84-kb genomic fragment in human chromosome 1 that encompasses the CFHR1 and CFHR3 genes represents a risk factor for hemolytic uremic syndrome (HUS) but has a protective effect in age-related macular degeneration (AMD). Here we identify CFHR1 as a novel inhibitor of the complement pathway that blocks C5 convertase activity and interferes with C5b surface deposition and MAC formation. This activity is distinct from complement factor H, and apparently factor H and CFHR1 control complement activation in a sequential manner. As both proteins bind to the same or similar sites at the cellular surfaces, the gain of CFHR1 activity presumably is at the expense of CFH-mediated function (inhibition of the C3 convertase). In HUS, the absence of CFHR1 may result in reduced inhibition of terminal complex formation and in reduced protection of endothelial cells upon complement attack. These findings provide new insights into complement regulation on the cell surface and biosurfaces and likely define the role of CFHR1 in human diseases.


Subject(s)
Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3b Inactivator Proteins/physiology , Complement Membrane Attack Complex/metabolism , Animals , Cells, Cultured , Complement C3-C5 Convertases/metabolism , Complement C3b/metabolism , Complement C3d/metabolism , Complement Factor H/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Erythrocytes/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Kidney/cytology , Kidney/metabolism , Microscopy, Confocal , Retina/cytology , Retina/metabolism , Sheep , Umbilical Veins/cytology , Umbilical Veins/metabolism
4.
Vaccine ; 26 Suppl 8: I9-14, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19388158

ABSTRACT

The complement system is a powerful part of the host innate immune defense and is aimed to damage and eliminate microbes and modified self-cells. To protect host cells and biological surfaces from damage mediated by complement activation products a tight control of the complement system is necessary. Imbalances in complement regulation contribute to tissue injury and can result in autoimmune diseases such as hemolytic uremic syndrome (HUS), membranoproliferative glomerulonephritis (MPGN) or age related macular degeneration (AMD). Disease associated mutations have been identified in several complement regulators or components, such as members of the factor H protein family. This group includes the major alternative pathway regulator, complement factor H (CFH) and five complement factor H related proteins (CFHR). Homozygous chromosomal deletion of a genomic 84 kb, chromosomal fragment which includes the genes CFHR1/CFHR3 is a risk factor for hemolytic uremic syndrome (HUS) at young age and is predominantly associated with the generation of autoantibodies to CFH, leading to a specific type of HUS, called DEAP (deficiency of CFHR and autoantibody positive)-HUS. The same deletion however is protective to the development of age related macular degeneration (AMD) in elderly people. Thus CFHR1 and CFHR3 proteins, and likely also the other members of this gene family are linked to human diseases. We here summarize the current knowledge about the role or association of CFHR1 and CFHR3 in the human diseases HUS and AMD.


Subject(s)
Blood Proteins/physiology , Complement C3b Inactivator Proteins/physiology , Hemolytic-Uremic Syndrome/etiology , Macular Degeneration/etiology , Complement Factor H/genetics , Hemolytic-Uremic Syndrome/genetics , Humans , Macular Degeneration/genetics
5.
J Immunol ; 165(7): 3999-4006, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11034410

ABSTRACT

Decay-accelerating factor (DAF or CD55) and membrane cofactor protein (MCP or CD46) function intrinsically in the membranes of self cells to prevent activation of autologous complement on their surfaces. How these two regulatory proteins cooperate on self-cell surfaces to inhibit autologous complement attack is unknown. In this study, a GPI-anchored form of MCP was generated. The ability of this recombinant protein and that of naturally GPI-anchored DAF to incorporate into cell membranes then was exploited to examine the combined functions of DAF and MCP in regulating complement intermediates assembled from purified alternative pathway components on rabbit erythrocytes. Quantitative studies with complement-coated rabbit erythrocyte intermediates constituted with each protein individually or the two proteins together demonstrated that DAF and MCP synergize the actions of each other in preventing C3b deposition on the cell surface. Further analyses showed that MCP's ability to catalyze the factor I-mediated cleavage of cell-bound C3b is inhibited in the presence of factors B and D and is restored when DAF is incorporated into the cells. Thus, the activities of DAF and MCP, when present together, are greater than the sum of the two proteins individually, and DAF is required for MCP to catalyze the cleavage of cell-bound C3b in the presence of excess factors B and D. These data are relevant to xenotransplantation, pharmacological inhibition of complement in inflammatory diseases, and evasion of tumor cells from humoral immune responses.


Subject(s)
Adjuvants, Immunologic/physiology , Antigens, CD/physiology , CD55 Antigens/physiology , Complement C3b Inactivator Proteins/physiology , Complement Pathway, Alternative/immunology , Membrane Glycoproteins/physiology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , CHO Cells , Complement C3 Convertase, Alternative Pathway , Complement C3-C5 Convertases , Complement C3b/immunology , Complement C3b/metabolism , Complement C3b/physiology , Complement Factor B/physiology , Cricetinae , Drug Synergism , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Humans , K562 Cells , Membrane Cofactor Protein , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Peptide Fragments/physiology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
6.
J Immunol ; 163(7): 3957-62, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10490997

ABSTRACT

C-reactive protein (CRP) is a major acute phase protein whose functions are not totally clear. In this study, we examined the interaction of CRP with factor H (FH), a key regulator of the alternative pathway (AP) of complement. Using the surface plasmon resonance technique and a panel of recombinantly expressed FH constructs, we observed that CRP binds to two closely located regions on short consensus repeat (SCR) domains 7 and 8-11 of FH. Also FH-like protein 1 (FHL-1), an alternatively spliced product of the FH gene, bound to CRP with its most C-terminal domain (SCR 7). The binding reactions were calcium-dependent and partially inhibited by heparin. In accordance with the finding that CRP binding sites on FH were distinct from the C3b binding sites, CRP preserved the ability of FH to promote factor I-mediated cleavage of C3b. We propose that the function of CRP is to target functionally active FH and FHL-1 to injured self tissues. Thereby, CRP could restrict excessive complement attack in tissues while allowing a temporarily enhanced AP activity against invading microbes in blood.


Subject(s)
C-Reactive Protein/physiology , Complement Activation/immunology , Complement Factor H/metabolism , Peptide Fragments/metabolism , Binding Sites/drug effects , Binding Sites/immunology , C-Reactive Protein/metabolism , Complement Activation/drug effects , Complement C3b Inactivator Proteins/physiology , Complement Factor H/genetics , Complement Factor H/physiology , Consensus Sequence/immunology , Heparin/pharmacology , Heparin, Low-Molecular-Weight/pharmacology , Humans , Peptide Fragments/immunology , Peptide Mapping , Protein Binding/drug effects , Protein Binding/immunology , Repetitive Sequences, Amino Acid/immunology , Surface Plasmon Resonance
8.
Transplantation ; 62(6): 729-34, 1996 Sep 27.
Article in English | MEDLINE | ID: mdl-8824468

ABSTRACT

As a potential source of organs for xenotransplantation, pigs that are transgenic for human decay accelerating factor (DAF) have been bred in order to overcome hyperacute rejection. We investigated the protective effect of human DAF in a porcine working heart model perfused by human blood. Hearts of normal landrace pits served as controls. The following parameters were measured: stroke work index, coronary flow and arteriovenous oxygen consumption, 6-keto prostaglandin F1alpha and prostaglandin E2 as markers of endothelial cell activation; creatine phosphokinase and lactate dehydrogenase for evaluation of the extent of myocardial damage; TNFalpha and IL-6 as markers of mononuclear cell activation. Histological and ultrastructural investigations from myocardial tissue sections were done at the end of perfusion. Human (h) DAF appeared to inhibit complement-mediated endothelial cell activation of transgenic pig hearts successfully. This was in contrast to landrace pig hearts, which had a sixfold increase of prostaglandin levels during perfusion with human blood. The cardiac weight increase during perfusion time due to interstitial edema tended to be less in the hDAF group. Myocardial damage was minimal in transgenic hearts, whereas normal pig hearts produced a threefold increase of creatine phosphokinase and lactate dehydrogenase levels. In these hearts, electron microscopy revealed single cell necrosis of myocytes and vacuolization of mitochondria with cristae rupture. According to the results obtained in the working heart model, the breeding of pigs that are transgenic for hDAF represents a promising step to making heart xenotransplantation a clinical reality in the future.


Subject(s)
Blood/immunology , CD55 Antigens/physiology , Complement C3b Inactivator Proteins/physiology , Graft Rejection/prevention & control , Heart Transplantation/immunology , Heart/physiology , Myocardium/immunology , Swine/immunology , Transplantation, Heterologous/immunology , Acute Disease , Animals , Animals, Genetically Modified , CD55 Antigens/genetics , Complement Activation , Complement C3b Inactivator Proteins/genetics , Female , Heart Function Tests , Humans , Interleukin-6/metabolism , Male , Perfusion , Species Specificity , Swine/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Mol Immunol ; 29(7-8): 957-64, 1992.
Article in English | MEDLINE | ID: mdl-1386142

ABSTRACT

RHP has been purified from the plasma of both normal individuals and patients with rheumatoid arthritis (RA). RHP from both these sources was shown to be identical with Factor H by reaction with antisera and N-terminal amino acid sequence analysis. Factor H, from both normal and RA sera, inhibited the solubilization of immune precipitates but did not affect prevention of immune precipitation. Factor H was shown to inhibit the haemolytic activity of fluid-phase C1, but unlike C1-inhibitor, it had little effect on C1 bound to EA (EAC1). Factor H was shown to complex with intact C1, to isolated C1q and to the C1r:C1s tetramer. However, binding of factor H to C1 did not dissociate the C1 macromolecule. A C1-Factor H complex was detected in the serum and plasma from normal individuals and patients with systemic lupus erythematosus and RA. Serum levels of this complex were reduced, by EDTA-treatment of serum and by activation of complement by the classical pathway.


Subject(s)
Arthritis, Rheumatoid/blood , Complement C1/physiology , Complement C3b Inactivator Proteins/physiology , Complement C1 Inactivator Proteins/pharmacology , Complement C3b Inactivator Proteins/isolation & purification , Complement Factor H , Humans , Macromolecular Substances , Molecular Weight , Protein Binding
10.
Biochem Biophys Res Commun ; 185(2): 505-9, 1992 Jun 15.
Article in English | MEDLINE | ID: mdl-1535192

ABSTRACT

The guinea pig complement factor H(FH) and the plasma precursor(PMCFS-1) of the major monocyte-chemotactic factor(MCFS-1) found in the skin site of delayed hypersensitivity reaction(DHR) induced in the guinea pigs were compared in the antigenicity and the function. Both anti-FH-IgG and anti-MCFS-1-IgG formed a single precipitation line against FH, PMCFS-1, MCFS-1 and guinea pig plasma, and these lines fused one another without any spur formation. The inhibition activity of FH for C3bBb was absorbed by anti-MCFS-1-F(ab')2 in a dose-dependent manner. PMCFS-1 inhibited C3bBb activity dose-dependently as FH. These results show that FH is identical to PMCFS-1 and imply that FH, converted to MCFS-1 plays as a monocyte-chemotactic factor in the site of DHR.


Subject(s)
Chemotactic Factors/physiology , Complement C3b Inactivator Proteins/physiology , Hypersensitivity, Delayed/physiopathology , Macrophages/physiology , Animals , Chemotactic Factors/immunology , Chemotaxis, Leukocyte , Complement C3b Inactivator Proteins/immunology , Complement Factor H , Guinea Pigs , Immunodiffusion , Protein Precursors/immunology
11.
J Am Soc Nephrol ; 2(8): 1328-37, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1385732

ABSTRACT

To determine the effects of surface-associated heparin on the capacity of hemodialysis membranes to activate complement, cellulose acetate (CA) membranes that were untreated and CA membranes that had been coated with heparin (HCA) were incubated with C3-depleted serum repleted with radio-labeled C3. Next, the proteins in the supernatant and those eluted from the membranes were subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis. C3 activation was quantified by determining the radioactivity of the C3a-containing band in the gel. Total C3a generation (fluid phase C3a plus membrane-associated C3a) was three times greater in the presence of HCA compared with CA. Most (88%) of the C3a generated in the presence of HCA, however, was adsorbed onto the membrane surface. Consequently, there was more C3a in the CA supernatant than in the HCA supernatant. To determine the mechanism by which heparin enhanced alternative pathway activity, binding studies with radiolabeled factor B and factor H were performed. HCA bound 3.4 times more factor B and 20 times more factor H than did CA. The binding of these proteins, however, was not dependent on complement activation. Studies designed to test the functional activity of isolated factor H and factor B that had been adsorbed to the membrane showed that factor H was active on both CA and HCA, whereas factor B was active only on HCA. These data demonstrate that heparin immobilized onto CA hemodialysis membrane enhances C3 activation but produces low levels of C3a in the fluid phase because of high surface adsorption of the anaphylatoxin. Heparin appears to augment alternative pathway activity by favoring the interactions of factor B with other constituents of the amplification C3 convertase of the alternative pathway of complement.


Subject(s)
Complement Pathway, Alternative/drug effects , Heparin/pharmacology , Membranes, Artificial , Renal Dialysis/instrumentation , Biocompatible Materials , Cellulose/analogs & derivatives , Complement C3/analysis , Complement C3a/analysis , Complement C3b Inactivator Proteins/physiology , Complement Factor B/physiology , Complement Factor H , Complement Pathway, Alternative/physiology , Humans
12.
Eur J Immunogenet ; 18(5-6): 399-403, 1991.
Article in English | MEDLINE | ID: mdl-1837734

ABSTRACT

The authors report the functional analysis of the purified mouse factor H allotypes H.1 and H.2, which were clearly distinguished from each other by an immunodiffusion test. Both allotypes acted as a co-factor for factor I in cleaving mouse C3b and we found no significant difference between their activities. The results strongly suggest that the function of mouse factor H for the co-factor activity has been well conserved between two allotypes.


Subject(s)
Complement C3b Inactivator Proteins/physiology , Serine Endopeptidases/physiology , Alleles , Animals , Complement C3b Inactivator Proteins/chemistry , Complement C3b Inactivator Proteins/genetics , Complement Factor H , Complement Factor I , Mice , Molecular Weight , Transfection
13.
Clin Exp Immunol ; 86 Suppl 1: 43-6, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1718643

ABSTRACT

CR1 and CR2 have served as unusual probes for the analysis of the two major functions of the immune system involving inflammation and the immune response, respectively. CR1, or some construct containing its active site SCRs, may find a role in the therapy of complement-dependent tissue injury, and may be used to define which diseases are caused by the inappropriate or excessive activation of this system. Although soluble forms of CR2 may be shown to have potential clinical utility when foreign antigen is given prospectively, as in monoclonal antibody therapy, perhaps the most important finding emanating from the analysis of this receptor is the recognition of a previously unrecognized membrane protein complex whose role in B cell development is yet to be determined. It is reasonable to predict that the function of the CD19 complex will be significant as it serves as the link between two evolutionarily distinct systems that share a common purpose of anti-microbial host defense.


Subject(s)
Complement Activation , Immunosuppressive Agents , Receptors, Complement/physiology , Animals , Antigens, CD/physiology , Antigens, Differentiation, B-Lymphocyte/chemistry , CD55 Antigens , Carrier Proteins/physiology , Complement C3b Inactivator Proteins/physiology , Complement Factor H , Integrin alphaXbeta2 , Membrane Cofactor Protein , Membrane Glycoproteins/physiology , Receptors, Complement/chemistry , Receptors, Complement 3b , Receptors, Complement 3d , Recombinant Proteins/administration & dosage , Solubility
14.
Mol Immunol ; 28(4-5): 375-82, 1991.
Article in English | MEDLINE | ID: mdl-1829503

ABSTRACT

Two proteins that are involved in cleavage of methylamine-treated C3 of guinea-pig origin (C3(MA)gp) have been isolated from guinea-pig serum. One of them functioned as a cofactor of human factor I (Ihu) for cleavage of C3(MA)gp and its molecular size was 150 kDa. The other was functionally pure and able to cleave C3(MA)gp together with human factor H (Hhu). They appear to be analogous to human factors H and I in the guinea-pig and will be referred to as Hgp and Igp. Methylamine-treated human C3 [C3(MA)hu] was not a compatible substrate for Hgp or Igp: little cleavage of C3(MA)hu was observed if human factor H (Hhu) or I was substituted with the guinea-pig counterpart. C3(MA)gp, on the other hand, served as a substrate, though less efficiently, for Hhu and Ihu. Human C4b-binding protein (C4bp) and membrane cofactor protein (MCP) as well as Hhu could participate in cleavage of C3(MA)gp by Igp or Ihu. In these assays, C3(MA)gp was degraded again less efficiently than C3(MA)hu. Interestingly, human C3b/C4b receptor (CR1) mediated factor I-dependent cleavage of C3(MA)hu and C3(MA)gp to a similar extent regardless the sources of factor I. These results suggest that factor I-dependent C3b regulatory system is species-specific except in the case of CR1, which may function as a cofactor irrespective of species.


Subject(s)
Complement C3b Inactivator Proteins/physiology , Complement C3b/physiology , Guinea Pigs/physiology , Serine Endopeptidases/physiology , Animals , Complement C3b Inactivator Proteins/chemistry , Complement C3b Inactivator Proteins/isolation & purification , Complement Factor H , Complement Factor I , Humans , In Vitro Techniques , Molecular Weight , Receptors, Complement/physiology , Receptors, Complement 3b , Serine Endopeptidases/chemistry , Serine Endopeptidases/isolation & purification , Species Specificity
15.
Eur J Immunol ; 20(12): 2643-50, 1990 Dec.
Article in English | MEDLINE | ID: mdl-1702720

ABSTRACT

Membrane regulatory molecules normally prevent complement activation by autologous cells, therefore we compared the membrane control system of human lymphoid cell lines which activate or not human complement through the alternative pathway (AP). Membrane expression of decay-accelerating factor (DAF), membrane cofactor protein (MCP), complement receptors (CR)1, CR2 and H was measured either by radioimmunoassay or enzyme-linked immunosorbent assay on cell lysates. Soluble extracts of isolated membranes were tested functionally for their ability to accelerate the decay of C3bBb C3-convertase and allow the cleavage of C3b by factor I. Both regulatory functions were detected in solubilized membranes of Ramos cells, which do not activate the AP, as well as on the potent AP activator, Raji. Raji cells were found to express CR2, DAF and MCP molecules, while MCP was the only known regulatory protein detected on Ramos cells which expressed neither CR1, nor CR2, H or DAF. The I-cofactor activity of both Raji and Ramos cells was immunoprecipitated by anti-MCP, but the decay-accelerating activity was not adsorbed by anti-DAF nor by any of the available antibodies. Two EBV genome-negative cell lines (BJAB, BL41) were tested before and after in vitro conversion by EBV. As previously shown, EBV-converted cell lines activate the AP more efficiently than EBV- cell lines. At the same time, EBV superinfection induces an increase of both AP regulatory functions of cell membranes and enhances the expression of DAF, MCP and CR2. The results of this study show that complement activation by lymphoid cell lines is not related to an impaired autologous control of these cells, but that the expression of regulatory molecules increases together with the appearance of activating structures on the cell surface. Our results also suggest the occurrence of a new factor involved in the decay-accelerating activity on BL lines.


Subject(s)
Antigens, CD , B-Lymphocytes/physiology , Complement Pathway, Alternative , Antigens, Differentiation, B-Lymphocyte/physiology , CD55 Antigens , Cell Line , Cell Membrane/physiology , Complement C3-C5 Convertases/metabolism , Complement C3b/metabolism , Complement C3b Inactivator Proteins/physiology , Complement Factor H , Complement Factor I , Herpesvirus 4, Human/immunology , Humans , In Vitro Techniques , Membrane Cofactor Protein , Membrane Glycoproteins/metabolism , Membrane Proteins/physiology , Receptors, Complement/physiology , Receptors, Complement 3b , Receptors, Complement 3d , Serine Endopeptidases/metabolism , Tumor Virus Infections/immunology
16.
Immunology ; 71(4): 598-600, 1990 Dec.
Article in English | MEDLINE | ID: mdl-1703989

ABSTRACT

Attachment of C3b to activators of the alternative pathway of complement results in a decrease in regulatory activity expressed by Factor H. Decay-accelerating factor (DAF) and Factor H were found to exhibit quantitatively similar decreases in regulatory activity toward the C3 convertase (C3b,Bb) bound to activators, such as zymosan (Zym) and rabbit erythrocytes (ER), compared to non-activators, such as sheep (ES) and bovine (EB) erythrocytes. Purified DAF and Factor H, in 0.1% NP-40, were assayed by measuring the amount required to release 50% of the radiolabelled Bb in 10 min from C3b,Bb on Zym or cross-linked erythrocytes. The relative effectiveness (i.e. the restriction index, RI) of DAF for accelerating the decay of C3b,Bb on the various particles was: ES (1.0), ER (0.04) and Zym (0.03). The RI for Factor H was: ES (1.0), ER (0.04) and Zym (0.07). The rate of decay of C3b,Bb induced by DAF and Factor H showed similar restriction. The results suggest that the regulatory properties of DAF are reduced if the cells on which it resides become activators of the alternative pathway as a result of transformation, virus infection or surface alteration. These findings may explain reports of dysfunctional DAF on alternative pathway-activating cells.


Subject(s)
Complement C3-C5 Convertases/metabolism , Complement C3b Inactivator Proteins/physiology , Complement Inactivator Proteins/physiology , Complement Pathway, Alternative/physiology , Membrane Proteins/physiology , Animals , CD55 Antigens , Complement Factor H , Erythrocytes/metabolism , Humans , Rabbits , Zymosan/metabolism
17.
Mol Immunol ; 27(9): 891-900, 1990 Sep.
Article in English | MEDLINE | ID: mdl-2145508

ABSTRACT

Kinetic experiments measuring the proteolytic activity of Bb and 33Kd fragment (the C-terminal domain of factor B) on C3 were performed in several conditions, in order to assess the role of factor B domains in the catalytic activity and magnesium binding. The experiments were carried out in fluid phase with 125I-C3 or C3(H2O) as substrates and in the presence of nonradioactive C3b as cofactor. The results indicate: (a) The C-terminal domain, 33Kd, possesses proteolytic activity on C3, which is Mg2(+)-independent, whereas proteolysis by Bb is enhanced in 5 mM Mg2+. (b) C3b behaves as cofactor of 33Kd proteolytic activity on C3 and factor H is able to inhibit this activity. (d) Kinetics of C3 proteolysis by 33Kd shows a lag phase which is also displayed by Bb in the absence but not in the presence of Mg2+. Taken together these data are consistent with the involvement of the N-terminal domain of Bb in Mg2+ binding, which results in an enhancement of the proteolytic activity on C3 of the adjacent C-terminal domain. A C3 convertase model accounting for these results is presented.


Subject(s)
Complement C3/metabolism , Complement Factor B/metabolism , Magnesium/metabolism , Complement C3b/physiology , Complement C3b Inactivator Proteins/physiology , Complement Factor B/chemistry , Complement Factor H , Humans , Peptide Fragments/metabolism
18.
Clin Exp Immunol ; 79(3): 454-8, 1990 Mar.
Article in English | MEDLINE | ID: mdl-2138528

ABSTRACT

We have investigated the development of substrate-based inhibitors of complement enzymes. Sequences around the scissile Arg77-Ser78 bond of C3 have been synthesized and tested as inhibitors of C3 convertase. The best inhibition was found with the tetrapeptide Ac-Arg-Ser-Asn-Leu-OH (H-576); extending this sequence in either direction reduced inhibitory activity. Preliminary experiments with peptides in which the scissile bond--CO--NH--was replaced with non-hydrolysable moieties such as--CO--CH2--(H-497) and--CH2--NH--(H-336) failed to show enhanced inhibition. One of the longer chain inhibitors H-416 containing DArg77-Ser78 was unexpectedly found to potentiate iC3 cleavage by factors I and H but did not inhibit the intact alternative pathway. The same peptide also bound to factor H. It is concluded that the binding requirements of the C3 convertase are more sophisticated than can be satisfactorily imitated simply by linear sequences around the scissile bond of C3.


Subject(s)
Complement Activating Enzymes/antagonists & inhibitors , Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3/metabolism , Oligopeptides/pharmacology , Complement C3b Inactivator Proteins/physiology , Complement Factor H
19.
Curr Top Microbiol Immunol ; 153: 147-62, 1990.
Article in English | MEDLINE | ID: mdl-2137073

ABSTRACT

While the mouse and human H proteins are structurally and functionally similar, they differ in their genetics. Whereas there is no evidence in humans for more than one gene; in mice the H locus is complex. Based on cDNA sequence and hybridization analysis of genomic cosmid clones, there are at least three distinct genes, all highly related to one another. The consensus repeating unit that comprises this molecule has obviously been duplicated numerous times, since it is present in many other molecules. Thus, it is not surprising to discover that there are several genes related to H in the mouse. A similar case has been described for two other members of this family. In humans, CR1 cDNA hybridizes to two distinct genomic clusters in the CR1 locus (Wong et al. 1989), and in mice, mCRY hybridizes to two regions in the genome, one on chromosome 1 and another on chromosome 8 (Aegerter-Shaw et al. 1987). It will be of interest to see if any other members of this family display as complex a genetic locus as murine H.


Subject(s)
Complement C3b Inactivator Proteins/physiology , Animals , Complement C3/physiology , Complement C3b Inactivator Proteins/genetics , Complement Factor H , DNA/genetics , Humans , Mice , Repetitive Sequences, Nucleic Acid , Sequence Homology, Nucleic Acid , Species Specificity
20.
Eur J Immunol ; 19(9): 1765-8, 1989 Sep.
Article in English | MEDLINE | ID: mdl-2529127

ABSTRACT

In addition to the 150-kDa factor H protein, we have previously described a 43-kDa factor H molecule in human plasma, which probably represents a translational product of the additional 1.8-kb mRNA for factor H. This factor H was isolated from human plasma by means of immunoaffinity chromatography and high-performance liquid chromatography. When tested for its functional activity, this purified 43-kDa H protein was shown to act as cofactor for factor I- mediated cleavage of fluid-phase C3b to iC3b.


Subject(s)
Complement Activation , Complement C3/metabolism , Complement C3b Inactivator Proteins/physiology , Complement Pathway, Alternative , Antibodies, Monoclonal/immunology , Blotting, Western , Complement C3b Inactivator Proteins/isolation & purification , Complement Factor H , Humans , Molecular Weight
SELECTION OF CITATIONS
SEARCH DETAIL
...