Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 51
1.
J Immunol ; 199(1): 292-303, 2017 07 01.
Article En | MEDLINE | ID: mdl-28533443

Factor H-related protein (FHR) 1 is one of the five human FHRs that share sequence and structural homology with the alternative pathway complement inhibitor FH. Genetic studies on disease associations and functional analyses indicate that FHR-1 enhances complement activation by competitive inhibition of FH binding to some surfaces and immune proteins. We have recently shown that FHR-1 binds to pentraxin 3. In this study, our aim was to investigate whether FHR-1 binds to another pentraxin, C-reactive protein (CRP), analyze the functional relevance of this interaction, and study the role of FHR-1 in complement activation and regulation. FHR-1 did not bind to native, pentameric CRP, but it bound strongly to monomeric CRP via its C-terminal domains. FHR-1 at high concentration competed with FH for CRP binding, indicating possible complement deregulation also on this ligand. FHR-1 did not inhibit regulation of solid-phase C3 convertase by FH and did not inhibit terminal complement complex formation induced by zymosan. On the contrary, by binding C3b, FHR-1 allowed C3 convertase formation and thereby enhanced complement activation. FHR-1/CRP interactions increased complement activation via the classical and alternative pathways on surfaces such as the extracellular matrix and necrotic cells. Altogether, these results identify CRP as a ligand for FHR-1 and suggest that FHR-1 enhances, rather than inhibits, complement activation, which may explain the protective effect of FHR-1 deficiency in age-related macular degeneration.


C-Reactive Protein/immunology , C-Reactive Protein/metabolism , Complement Activation , Complement C3b Inactivator Proteins/immunology , Complement C3b Inactivator Proteins/metabolism , Binding Sites , C-Reactive Protein/chemistry , C-Reactive Protein/pharmacology , Complement C3-C5 Convertases , Complement C3b/immunology , Complement C3b/pharmacology , Complement C3b Inactivator Proteins/pharmacology , Complement Factor H , Extracellular Matrix/drug effects , Extracellular Matrix/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Ligands , Macular Degeneration/immunology , Protein Binding , Serum Amyloid P-Component/immunology , Serum Amyloid P-Component/metabolism
2.
Invest Ophthalmol Vis Sci ; 50(3): 1392-9, 2009 Mar.
Article En | MEDLINE | ID: mdl-19029031

PURPOSE: Studies implicate activation of complement among the processes involved in the pathogenesis of age-related macular degeneration (AMD). Questions pertain to the trigger(s) responsible for the complement-associated events. The authors previously reported that photooxidation products of A2E can activate complement. Here they have further explored these events. METHODS: In vitro assays using human serum as a source of complement were used, and the C3 split product iC3b was measured by enzyme immunoassay. Serum was placed in contact with ARPE-19 cells and polarized human fetal retinal pigment epithelium that had accumulated A2E and were irradiated (430 nm). Serum was also incubated in wells precoated with bisretinoid pigments of lipofuscin and their oxidized forms. iC3b generation in normal human serum (NHS) was compared with that in factor B-depleted and C1q-depleted human serum. RESULTS: iC3b levels were elevated in NHS placed in contact with A2E-laden retinal pigment epithelium that were irradiated to generate A2E photooxidation products. iC3b was also increased in serum incubated in wells precoated with peroxy-A2E, the lipofuscin pigment all-trans-retinal dimer, and oxidized forms of all-trans-retinal dimer. Substitution of NHS with factor B-depleted sera abrogated these increases in iC3b. Complement activation was also suppressed by the addition of C-reactive protein and by a C3 cleavage inhibitor. CONCLUSIONS: The authors suggest that bisretinoid pigments of retinal pigment epithelial lipofuscin, subsequent to photoactivation and cleavage, serve to activate complement. Complement activation by this mechanism is dependent on the alternative pathway and can be modulated by an inhibitor of C3 cleavage. These events in the setting of complement dysregulation could contribute to the chronic inflammation that underlies AMD pathogenesis.


Complement Activation/physiology , Complement C3b/biosynthesis , Lipofuscin/metabolism , Pyridinium Compounds/metabolism , Retinal Pigment Epithelium/metabolism , Retinoids/metabolism , Apoptosis/drug effects , C-Reactive Protein/pharmacology , Cells, Cultured , Complement C3b/antagonists & inhibitors , Complement C3b Inactivator Proteins/pharmacology , Complement Pathway, Alternative/physiology , Etoposide/pharmacology , Humans , Immunoenzyme Techniques , Lipofuscin/chemistry , Oxidation-Reduction , Peptides, Cyclic/pharmacology , Pyridinium Compounds/chemistry , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/embryology , Retinoids/chemistry
3.
Biochem Pharmacol ; 57(12): 1439-46, 1999 Jun 15.
Article En | MEDLINE | ID: mdl-10353266

Rosmarinic acid has been reported to inhibit complement activation in vivo as well as in vitro. Previous studies suggested that the inhibitory effect was due to inhibition of C3/C5 convertases, but inhibition of C3b attachment would yield the same results. Recent work in our laboratory demonstrated that compounds with polyhydroxylated phenyl rings are highly reactive with the thioester bond in nascent C3b. These compounds block complement activation by preventing attachment of C3b to the activating surface. Because rosmarinic acid contains two 3,4-dihydroxyphenyl groups, the current study was undertaken to re-examine the mechanism of inhibition by analyzing the effect of rosmarinic acid on C3b attachment. In assays using purified complement proteins, rosmarinic acid inhibited covalent attachment of C3b to cells with an 1C50 = 34 microM. Inhibition of C5 convertase activity required 1500 microM rosmarinic acid, and no significant inhibition of the C3 convertase enzyme, which produces C3b from C3, was observed at 10,000 microM. In hemolytic assays using human serum, rosmarinic acid was shown to inhibit activation of both the classical (IC50 = 180 microM) and the alternative (IC50 = 160 microM) pathways of complement. Rosmarinic acid concentrations up to 10,000 microM did not cause direct inactivation of C3. Radioiodination of rosmarinic acid was used to demonstrate covalent activation-dependent incorporation of rosmarinic acid specifically into the thioester-containing alpha'-chain of nascent C3b. These findings indicate that inhibition of complement activation by rosmarinic acid is due to the reaction of rosmarinic acid with the activated thioester of metastable C3b, resulting in covalent attachment of the inhibitor to the protein.


Cinnamates/pharmacology , Complement C3b Inactivator Proteins/pharmacology , Complement C3b/drug effects , Animals , Cinnamates/chemistry , Complement Activation , Complement C3/antagonists & inhibitors , Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3-C5 Convertases/metabolism , Depsides , Erythrocytes/drug effects , Hemolysis/drug effects , Humans , Sheep , Rosmarinic Acid
4.
Biochem Pharmacol ; 51(6): 797-804, 1996 Mar 22.
Article En | MEDLINE | ID: mdl-8602875

An intramolecular thioester bond in complement protein C3 is vital for covalent attachment of C3b (the proteolytically activated form of C3) to biological surfaces and for activation of the complement system. Proteolytic removal of C3a from C3 activates the thioester in the C3b fragment. Activated C3b primarily forms ester bonds with hydroxyl groups of carbohydrates on complement activating surfaces, but it has also been shown to react with the hydroxyl group of tyrosine and with specific Ser and Thr residues on IgG and on complement protein C4b. To examine the reactivity of the thioester, several families of hydroxylated compounds were examined. Reactivity of a series of substituted phenols varied over two orders of magnitude and demonstrated a linear correlation between reactivity and the Hammett substituent constants. Hydroxylated drugs including members of the L-DOPA/epinephrine family and hydroxamic acids also were examined. Compounds were identified that were 20,000 times more reactive than carbohydrates. These compounds were found to inhibit both the classical and alternative pathways of complement activation. Although the specificity of the thioester for its natural biological targets appears to be determined by many structural features, the data presented here demonstrate that increasing the nucleophilic character of the target hydroxyl group can increase the potency of a synthetic inhibitor many orders of magnitude.


Complement C3/metabolism , Complement C3b Inactivator Proteins/pharmacology , Complement C3b/metabolism , Humans , Hydroxylation , Phenols/metabolism , Phenols/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
5.
J Invest Dermatol ; 104(2): 297-301, 1995 Feb.
Article En | MEDLINE | ID: mdl-7829888

The accumulation of polymorphonuclear leukocytes (PMN) beneath the stratum corneum is a characteristic histopathologic finding in various aseptic pustular dermatoses. To elucidate the pathomechanism involved in this phenomenon, we investigated whether PMN also attach to a sheet of corneocytes in vitro. A 1-cm2 corneocyte sheet was attached to a sterile glass slide with double adhesive tape used for skin graft surgery before incubating with human serum. The PMN suspension then was applied to the sheet. Attached cells were stained with May-Grunwald-Giemsa and counted with a computer image analyzer. We quantitatively assessed PMN adhesion to the serum-treated corneocyte sheets, which was mediated by activation of the alternative complement pathway. Addition of either anti-CD18 or anti-CD11b antibody to the assay system resulted in a marked reduction of PMN adhesion. We also demonstrated immunohistochemically that iC3b was formed on the serum-treated corneocytes. These findings suggest that PMN attach to serum-treated corneocytes through an interaction of CR3 expressed on PMN with iC3b-coated corneocytes. In addition, we found that this adhesion was enhanced by activation of PMN with phorbol myristate acetate. From these results, we speculate that complement activation by corneocytes occurs in the cutaneous lesions of aseptic pustular dermatoses and that PMN can be stimulated by the interaction with iC3b-opsonized corneocytes as well as by chemotaxins, leading to damage of the surrounding epidermal keratinocytes.


Blood Physiological Phenomena , Neutrophils/cytology , Skin/cytology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Complement Activation , Complement C3b Inactivator Proteins/pharmacology , Humans , Tetradecanoylphorbol Acetate/pharmacology
6.
Tohoku J Exp Med ; 174(3): 269-77, 1994 Nov.
Article En | MEDLINE | ID: mdl-7761992

The beta-amyloid protein deposits of Alzheimer disease, whether in diffuse or consoliated form, are an agglomeration of many extracellular proteins. At least 35 have been reported as components of senile plaques, most of which also occur in diffuse deposits. More than half of these proteins are directly associated with the immune system. Since diffuse deposits are believed to be the precursors of senile plaques, it is important to define the precise molecular events that lead to the transition. Diffuse deposits share with senile plaques the presence of opsonizing components of complement, the complement activators beta-amyloid protein, amyloid P, thrombin, and apolipoprotein E. However, senile plaques contain, in addition, dystrophic neurites, agglomerates of activated microglia, components of the membrane attack complex, and the inhibitors of the membrane attack complex, clusterin, protectin and vitronectin. Microglial cells are professional phagocytes which possess the respiratory burst apparatus when activated. It produces extracellular superoxide molecules which can then form additional toxic products such as hydrogen peroxide and hydroxyl free radicals. It has long been known that opsonized zymosan is a powerful activator of the respiratory burst system. We found this activation could be inhibited by antibodies to complement receptors in the nanomolar range. Dapsone and indomethacin, two antiinflammatory agents that may have therapeutic potential in Alzheimer disease, were weakly inhibitory (10(-4) M range).(ABSTRACT TRUNCATED AT 250 WORDS)


Alzheimer Disease/pathology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain Chemistry , Complement Pathway, Classical , Microglia/pathology , Models, Neurological , Nerve Tissue Proteins/analysis , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid/chemistry , Amyloid beta-Peptides/analysis , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Complement C3b Inactivator Proteins/pharmacology , Complement C3b Inactivator Proteins/therapeutic use , Complement Membrane Attack Complex/analysis , Complement System Proteins/analysis , Extracellular Matrix Proteins/analysis , Humans , Microglia/chemistry , Microglia/immunology , Neurites/pathology , Opsonin Proteins/immunology , Respiratory Burst/drug effects , Zymosan/antagonists & inhibitors , Zymosan/immunology
7.
J Periodontal Res ; 28(5): 346-53, 1993 Sep.
Article En | MEDLINE | ID: mdl-8410599

Anaerobic microorganisms in periodontal pockets produce toxic amounts of hydrogen sulfide. The capacity of polymorphonuclear leukocytes to kill a capsulated and a non-capsulated variant of a group B streptococcal strain was studied in presence and absence of sulfide. The killing was equally efficient under aerobic and anaerobic conditions. However, in presence of sulfide the killing of the capsulated variant of the strain was significantly inhibited. Since this strain required higher serum concentrations to be killed by the polymorphonuclear leukocytes, it suggested that sulfide interfered with the opsonization of the bacteria. The capacity of sulfide to split the disulfide bonds of complement factor 3 and immunoglobulin G, deposited on the bacterial surface, was evaluated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. There was no detectable effect of 2 mM sulfide on immunoglobulin G. However, sulfide released from opsonized bacteria the beta-chain of C3b C3bi, and the C-terminal part of the alpha-chain of C3bi. This region of the alpha-chain of C3bi has been suggested to bind to the complement receptor 3 of polymorphonuclear leukocytes. The beta-chain of C3b/C3bi may augment the binding of opsonized bacteria to the complement receptors of polymorphonuclear leukocytes. The formation of sulfide by the microflora of the periodontal pockets may provide conditions for the bacteria to escape important parts of the host immune system.


Complement C3b/drug effects , Neutrophils/physiology , Phagocytosis/drug effects , Streptococcus agalactiae/drug effects , Sulfides/pharmacology , Anaerobiosis , Blood Bactericidal Activity/drug effects , Complement C3b/chemistry , Complement C3b Inactivator Proteins/pharmacology , Disulfides/chemistry , Electrophoresis, Polyacrylamide Gel , Humans , Immunoglobulin G/drug effects , Neutrophils/immunology , Opsonin Proteins/drug effects , Opsonin Proteins/physiology , Streptococcus agalactiae/immunology , Streptococcus agalactiae/metabolism , Sulfides/metabolism
8.
Mol Immunol ; 30(10): 855-64, 1993 Jul.
Article En | MEDLINE | ID: mdl-8341280

Covalent Superose microspheres-bound C3b was used as a model system to simplify the analysis of antigen-bound C3b modifications during antigen processing. The model was set up using purified C3 and Superose-bound trypsin. C3b was covalently bound to Superose through an ester link, as indicated by lability to hydroxylamine treatment at alkaline pH. C3b-Superose was incubated with L subcellular fraction, enriched in endosomes/lysosomes, purified from U937 cell line. Two types of limited activities on the C3b-Superose model system were detected: (i) a proteolytic activity cleaving C3b into mainly a C3c-like fragment which was released and a C3d-like fragment of apparent M(r) 32 kDa which remained bound to Superose through the original ester link; (ii) an esterolytic activity cleaving the ester bond and releasing C3b. Inhibition experiments pointed to the involvement of serine, aspartyl and cysteine proteases. Cathepsin B appeared most probably as one of the major proteases of L fraction catalysing the proteolysis of the C3b-bound. Kinetic studies were in favour of a good stability on the ester bond, supporting an effective role of C3b as a chaperone during the extracellular and intracellular travel of C3b-bound antigen.


Complement Activation/physiology , Complement C3b/metabolism , Endopeptidases , Cathepsin B/metabolism , Cathepsin D/metabolism , Cathepsin L , Cathepsins/metabolism , Complement Activation/drug effects , Complement C3b/drug effects , Complement C3b Inactivator Proteins/pharmacology , Cysteine Endopeptidases , Electrophoresis, Polyacrylamide Gel , Enzyme Precursors/pharmacology , Humans , Hydrogen-Ion Concentration , Hydrolysis , Immunoblotting , Lysosomes/metabolism , Microspheres , Time Factors , Tumor Cells, Cultured
9.
Cancer Res ; 53(3): 592-9, 1993 Feb 01.
Article En | MEDLINE | ID: mdl-8425193

The molecular mechanism of complement resistance of the human SK-MEL-170 melanoma cell line was investigated. The cells have been shown to express the C3b-cleaving membrane protease p65. To delineate the molecular consequences of the C3b-cleaving activity for the complement cytotoxicity, the molecular events during the initiation (R24 monoclonal antibody, C1), amplification (C4, C3), and membrane attack (C5, C9) phases of complement were studied in comparison to a complement-susceptible human melanoma line (SK-MEL-93-2). No cleavage of C4b and C5b, 2 molecules structurally similar to C3b, was observed on the cells during classical pathway activation indicating the specificity of the p65 protease for the C3b molecule. The rapid degradation of C3b by p65 on the surface of complement-resistant SK-MEL-170 cells generates a M(r) 30,000 C3 alpha'-chain-fragment detectable as early as 1 min after complement activation, whereas no such fragment was present in detectable amounts on complement-susceptible cells. As a result of the rapid C3b proteolysis by p65 on resistant SK-MEL-170 cells, less C5 convertases are formed, which in turn results in the formation of a lower number of terminal complement components and membrane attack complexes. R24 antibody and C1q binding to the resistant cells was slightly lower as to susceptible cells. C4 binding studies, however, revealed that the observed difference in antibody and C1q binding has no influence on the complement resistance of SK-MEL-170 cells: significantly more C4b was bound to complement-resistant (1565 +/- 92 fg/cell) as compared to susceptible cells (715 +/- 31 fg/cell). On extraction of the molecular forms of C4 bound to the cell membranes, an additional high molecular weight C4 species--apparently a C4b-C4b homodimer--appeared only on the resistant SK-MEL-170 cells that may function as a residual back-up C5 convertase. Collectively, these results show that SK-MEL-170 human melanoma cells evade complement-mediated cytolysis despite sufficient activation of early components of the classical complement pathway by p65-mediated rapid degradation of surface-bound C3b, leading to a significant reduction in membrane attack complex formation. Thus, rapid cleavage of surface deposited C3b was established as a powerful mechanism of complement resistance.


Complement Activation/physiology , Complement C3b/metabolism , Complement System Proteins/immunology , Endopeptidases/physiology , Melanoma/enzymology , Animals , Antibodies, Monoclonal/metabolism , Cell Membrane/enzymology , Complement C1q/metabolism , Complement C3/metabolism , Complement C3/physiology , Complement C3b/physiology , Complement C3b Inactivator Proteins/pharmacology , Complement C4/metabolism , Complement C4/physiology , Complement C5/metabolism , Complement C5/physiology , Complement C9/metabolism , Complement C9/physiology , Complement Membrane Attack Complex/physiology , Humans , Melanoma/metabolism , Melanoma/physiopathology , Mice , Mice, Inbred BALB C , Tumor Cells, Cultured
10.
Eur J Immunol ; 21(4): 967-72, 1991 Apr.
Article En | MEDLINE | ID: mdl-1826890

We studied the capacity of human factor H to promote the secretion of a lymphocyte-activating factor (LAF) by human monocytes cultured under serum-free conditions. Presence of LAF in the culture supernatants was assessed with the mouse thymocyte assay. Highly purified factor H alone had no effect on thymocyte proliferation. When monocytes were cultured with factor H for 24 h, a significant secretion of LAF was observed. The effect was dose dependent over a range of factor H concentrations from 1 to 15 micrograms/ml. Polymyxin B did not abrogate the capacity of factor H to induce LAF secretion. Adsorption of factor H preparations onto anti-factor H-Sepharose completely suppressed the phenomenon. Conversely, the activity was recovered in the acidic eluate. Furthermore, factor H subpopulation phi 2, that was able to bind to phenyl-Sepharose, was a stronger inducer of LAF secretion by monocytes than the subpopulation phi 1 (which did not bind to phenyl-Sepharose). Using a specific radioimmunoassay for interleukin 1-beta (IL 1 beta), we observed a good correlation between the LAF activity and the amount of IL 1 beta secreted by human monocytes stimulated with factor H. We have shown previously that factor H (phi 2) bound specifically on Raji cells whereas factor H (phi 1) did not. These results argue for the participation of the interaction of factor H with its receptor to stimulate the secretion of IL 1 by monocytes and that the phi 2 form of factor H is a ligand for the human factor H receptor.


Complement C3b Inactivator Proteins/pharmacology , Interleukin-1/metabolism , Monocytes/drug effects , Cells, Cultured , Complement C3b Inactivator Proteins/chemistry , Complement Factor H , Humans , Monocytes/metabolism
11.
Science ; 249(4965): 146-51, 1990 Jul 13.
Article En | MEDLINE | ID: mdl-2371562

The complement system is an important mediator of the acute inflammatory response, and an effective inhibitor would suppress tissue damage in many autoimmune and inflammatory diseases. Such an inhibitor might be found among the endogenous regulatory proteins of complement that block the enzymes that activate C3 and C5. Of these proteins, complement receptor type 1 (CR1; CD35) has the most inhibitory potential, but its restriction to a few cell types limits its function in vivo. This limitation was overcome by the recombinant, soluble human CR1, sCR1, which lacks the transmembrane and cytoplasmic domains. The sCR1 bivalently bound dimeric forms of its ligands, C3b and methylamine-treated C4 (C4-ma), and promoted their inactivation by factor I. In nanomolar concentrations, sCR1 blocked complement activation in human serum by the two pathways. The sCR1 had complement inhibitory and anti-inflammatory activities in a rat model of reperfusion injury of ischemic myocardium, reducing myocardial infarction size by 44 percent. These findings identify sCR1 as a potential agent for the suppression of complement-dependent tissue injury in autoimmune and inflammatory diseases.


Autoimmune Diseases/immunology , Complement Inactivator Proteins/pharmacology , Myocardial Reperfusion Injury/immunology , Myocardium/pathology , Receptors, Complement/pharmacology , Animals , Autoimmune Diseases/pathology , Complement Activation , Complement C3/antagonists & inhibitors , Complement C3b Inactivator Proteins/pharmacology , Complement C4b/antagonists & inhibitors , Complement C5/antagonists & inhibitors , Complement Inactivator Proteins/ultrastructure , Disease Models, Animal , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Necrosis , Rats , Receptors, Complement/ultrastructure , Recombinant Proteins/pharmacology
12.
J Pediatr ; 116(5): S92-7, 1990 May.
Article En | MEDLINE | ID: mdl-2139466

The third component of complement, the central protein of the complement cascade, occurs in two principal allotypes, C3S and C3F. An excess frequency of the F allotype has been implicated in a number of disease states, including some forms of glomerulonephritis. These associations have been explained by functional differences between C3S and C3F. We examined several complement functions, using purified preparations of C3S or C3F. The C3S allotype was 1.3 times more efficient than C3F in a hemolytic assay employing sensitized sheep erythrocytes; this difference was shown to arise from a slightly more efficient deposition of C3F on the cell surface. These differences are trivial and of much less magnitude than the functional differences between C4A and C4B. There were no differences between allotypes in their ability to be converted to inactive C3b (C3bi) by complement factors H and I or by CR1 and factor I. No significant differences were seen between the allotypes and their ability to support solubilization of preformed immune complexes.


Complement C3/genetics , Polymorphism, Genetic , Animals , Antigen-Antibody Complex/genetics , Blood Proteins/analysis , Cattle , Complement C3/analysis , Complement C3b Inactivator Proteins/pharmacology , Complement C4/analysis , Complement Factor H , Complement Factor I , Erythrocytes/immunology , Hemolysis/genetics , Humans , Phenotype , Rabbits , Serine Endopeptidases/pharmacology , Sheep , Solubility
13.
Complement Inflamm ; 7(1): 30-41, 1990.
Article En | MEDLINE | ID: mdl-2138950

The reaction of radiolabeled C3b-binding proteins with C3b-coated particles has been investigated. CR1 binding was inhibited by factor H and factor B (in the presence of properdin), but not by properdin alone. CR2 and MCP binding were also inhibited by factor H. Therefore factor H, factor B, CR1, CR2 and MCP probably comprise a group of mutually competitive proteins with similar or overlapping binding sites on C3b. These results correlate with their structural homology and suggest that they all evolved from a single C3b-binding molecule. Factor H, CR1 and MCP are also cofactors for the factor-I-mediated cleavage of C3b. A species incompatibility between rat factor I and human CR1 for the cleavage of human C3b suggests the possibility that cofactors may also function by interacting directly with factor I.


Antigens, CD , Complement C3b Inactivator Proteins/metabolism , Complement C3b/metabolism , Complement Factor B/metabolism , Enzyme Precursors/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Animals , Binding Sites , Binding, Competitive , Complement C3b Inactivator Proteins/pharmacology , Complement Factor B/pharmacology , Complement Factor H , Humans , Membrane Cofactor Protein , Properdin/pharmacology , Rats , Sheep
14.
Immunology ; 68(4): 445-8, 1989 Dec.
Article En | MEDLINE | ID: mdl-2532618

IgG mouse monoclonal antibody (mAb) was prepared by fusion of spleen cells from mice immunized with human C3d (mAb:C3d) using syngeneic thymocytes as feeder cells. mAb:C3d was assessed for its effect on the stabilization of the cell-bound alternative pathway C3 convertase EAC3bBb. It bound to cell-bound C3b and stabilized C3bBb at 30 degrees in the presence of EDTA-GVB. The plasma protein H reduced the stabilization effect of the stabilized C3 convertase. These results suggest that binding of antibody to C3d may stabilize C3bBb. It seems likely that such antibody induces in C3b conformational change, which increases the C3bBb complex stability.


Antibodies, Monoclonal/pharmacology , Complement Activating Enzymes/immunology , Complement Activation , Complement C3-C5 Convertases/immunology , Complement C3d/immunology , Complement Pathway, Alternative , Complement C3 Nephritic Factor/pharmacology , Complement C3b Inactivator Proteins/pharmacology , Complement Factor H , Enzyme Stability , Humans , Immunoglobulin G/pharmacology
15.
Mol Immunol ; 26(10): 939-48, 1989 Oct.
Article En | MEDLINE | ID: mdl-2531841

C3f, a heptadeca-peptide having the amino acid sequence of NH2-Ser-Ser-Lys-Ile-Thr-His-Arg-Ile-His-Trp-Glu-Ser-Ala-Ser-Leu-Leu-Arg- COOH, is liberated during the catabolic degradation of C3b in serum. The amino acid sequence of C3f is known both from the cDNA-derived structure of C3 and from protein analysis after isolation of the natural factor. C3f was synthesized by solid phase peptide synthesis. Both natural and synthetic C3f had identical retention times by RP-18 high performance liquid chromatography (HPLC) analysis and the respective amino acid compositions agreed with the expected theoretical values. C3f, but not des-Arg-C3f, was weakly spasmogenic inducing contraction of guinea pig ileum at a level of 5-10 x 10(-6) M. Since C3f and C3a were cross-tachyphylactic, it was concluded that these two spasmogens compete for the same receptors. Both C3f and des-Arg-C3f at concns of 1-4 x 10(-4) M enhanced vascular permeability in guinea pig skin. These observations further suggest that C3f functionally resembles C3a anaphylatoxin. Formation of C3f in human serum following CVF activation of C3 could be demonstrated by radioimmunoassay (RIA). Digestion of C3f with purified human serum carboxypeptidase N produced C3f-desArg. These observations suggest that when serum complement protein C3 undergoes conversion to C3b, further degradation by Factors H and I readily generates C3f. C3f is a weak spasmogen that functions like C3a anaphylatoxin and C3f-desArg is a major metabolite in serum.


Complement C3/physiology , Complement C3b Inactivator Proteins/pharmacology , Complement C3b/metabolism , Serine Endopeptidases/pharmacology , Amino Acid Sequence , Animals , Capillary Permeability , Chromatography, High Pressure Liquid , Complement C3/isolation & purification , Complement C3/metabolism , Complement Factor H , Complement Factor I , Elapid Venoms/pharmacology , Guinea Pigs , Humans , Molecular Sequence Data , Muscle Contraction , Radioimmunoassay
16.
J Immunol ; 142(8): 2771-7, 1989 Apr 15.
Article En | MEDLINE | ID: mdl-2522969

Tumor cells have adapted several strategies which permit them to grow in an immunologically hostile environment. The C system can potentially destroy these cells; however, its action needs to be specifically potentiated on the surface of the tumor cells. To this end, a heteroconjugate composed of a mouse mAb and of the human C3b C component has been generated by using the heterobifunctional reagent N-succinimidyl-3-(2-pyridyldithio)propionate. The two mAb which were used in this study are V1-10 and TIB219 which bind to the human and mouse transferrin receptors, respectively. The mAb-C3b conjugates were purified by gel filtration and were each composed of one mAb and one C3b. They bound to the human K562 and HL60 or mouse ALB1 cell lines and amplified the killing of these cells by C from 10 to 15% to 70 to 100%. Fresh normal human or mouse sera were used as a source of C. The mAb-C3b conjugates activated primarily the alternative pathway of C since only C3 and factor B but not C4 were cleaved in the sera. After disulfide-linking to the mAb, the C3b became highly resistant to inactivation by factors H and I, probably due to its reduced factor H binding capacity. On the other hand, the conjugated C3b bound factor B better than free C3b and produced more C3 convertases which expressed increased stability. These results suggest that mAb-C3b conjugates may serve as an effective tool for the specific activation of the cytolytic C system on selected cells. As such, they may be used in vitro or in vivo to target the autologous C to tumor cells or to lymphocytes and may promote tumor immunotherapy.


Antibodies, Monoclonal/administration & dosage , Complement C3b/administration & dosage , Cytotoxicity, Immunologic , Neoplasms/immunology , Animals , Complement C3-C5 Convertases/pharmacology , Complement C3b/metabolism , Complement C3b Inactivator Proteins/metabolism , Complement C3b Inactivator Proteins/pharmacology , Complement Factor H , Complement Pathway, Alternative , Elapid Venoms/administration & dosage , Humans , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/therapy , Tumor Cells, Cultured
18.
J Immunol ; 140(5): 1581-7, 1988 Mar 01.
Article En | MEDLINE | ID: mdl-3346542

Liposomes as defined model membranes were used to quantitatively study the effects of specific sialic acid containing glycolipids on activation of the alternative pathway of human C. Liposomes containing dimyristoylphosphatidylethanolamine, cholesterol, and cerebrosides at molar ratios of 1.0/0.75/0.33 activated the alternative pathway in human serum treated with MgEGTA. Activation was measured by C3 conversion and the deposition of total C3 and functional C3b on the liposome surface. The monosialoganglioside GM1, when incorporated into the activating liposome membrane at molar ratios between 10(-5) and 10(-2), inhibited activation in a dose-dependent manner. Sialosylparagloboside also inhibited activation in human serum, and inhibition was completely reversed after neuraminidase treatment. The degree of inhibition by GM1 correlated with the relative amount of GM1 exposed on the liposome surface. Sialic acid did not directly inhibit the binding of C3b when liposomes containing gangliosides were incubated with the purified components C3, B, D, and P. GM1 did inhibit activation when liposomes were incubated with a mixture of purified C3, B, D, P, H, and I. Binding assays with radiolabeled H showed increased binding of H to liposome-bound C3b in the presence of GM1. These results establish the ability of sialic acid on glycolipids to promote H binding to C3b and thereby regulate alternative pathway activation on a defined lipid membrane.


Complement Activation/drug effects , Complement C3b Inactivator Proteins/pharmacology , Complement Pathway, Alternative/drug effects , Gangliosides/pharmacology , Binding, Competitive , Complement C3/metabolism , Complement Factor B/metabolism , G(M1) Ganglioside/pharmacology , Globosides/pharmacology , Humans , Lipid Bilayers/metabolism , Liposomes , Models, Biological , Neuraminidase/pharmacology
19.
J Immunol ; 140(5): 1588-94, 1988 Mar 01.
Article En | MEDLINE | ID: mdl-3346543

Liposomes were used to determine whether gangliosides containing certain structurally defined analogues of sialic acid could inhibit activation of the alternative pathway of human C. Gangliosides containing sialic acid residues with modifications in the N-acetyl group, carboxyl group, or polyhydroxylated tail were either isolated from natural sources or prepared by chemical modification of the native sialic acid structure. Sialic acid lost more than 90% of its inhibitory activity after removal of just the C9 carbon from the polyhydroxylated tail. Sialic acid was also unable to inhibit activation after converting the carboxyl group to a hydroxymethyl group. Galactose oxidase/NaB3H4 treatment of liposomes containing gangliosides with native or modified sialic acid residues confirmed that neither modification altered the amount of gangliosides exposed at the liposome surface. Changing the N-linked acetyl group to a glycolyl group had no effect on the inhibitory activity of sialic acid. These data further define the structural features of sialic acid that are important in regulation of alternative pathway activation. Both the C9 carbon of the polyhydroxylated tail and the carboxyl group are essential for this function; whereas, the N-linked acetyl group may be modified without loss of activity.


Complement Activation/drug effects , Complement Pathway, Alternative/drug effects , G(M1) Ganglioside/pharmacology , Sialic Acids/pharmacology , Binding, Competitive , Borohydrides , Carbohydrate Conformation , Complement C3b Inactivator Proteins/pharmacology , Humans , Hydroxylation , Liposomes , N-Acetylneuraminic Acid , Structure-Activity Relationship
20.
J Immunol ; 134(5): 3332-8, 1985 May.
Article En | MEDLINE | ID: mdl-3156931

A radioimmunoassay with the use of soluble 125I-Fab monoclonal anti-CR1 and rabbit IgG anti-CR1 bound to Staphylococcus aureus particles was employed to detect and quantitate CR1 antigen in human plasma. Among 16 normal individuals the concentration of soluble CR1 in plasma ranged from 13 to 81 ng/ml, and a similar range of concentration was found in plasma from 15 patients having systemic lupus erythematosus (SLE). The amount of plasma CR1 in normal donors, but not in SLE patients, significantly correlated with the number of CR1 sites on erythrocytes (r = 0.90, p less than 0.001), and was 7.1% of the amount of receptor that was present on erythrocytes in blood. The concentration of soluble CR1 was not diminished by ultracentrifugation or ultrafiltration of plasma, was not affected by various modes of anti-coagulation or even by clotting of blood, and did not change during incubation of blood at 4 degrees C for up to 4 hr. On sucrose density gradient ultracentrifugation of plasma the CR1 was distributed as a broad peak that overlapped the plasma protein profile. The Mr of plasma CR1 was identical to that of erythrocyte CR1 when assessed by polyacrylamide gel electrophoresis in the presence of sodium dodecyl sulfate and immunoblotting. In addition, the plasma form of CR1 exhibited the same structural phenotype as did receptor from erythrocytes of the same individual. CR1 antigen purified from plasma was as active as CR1 from erythrocytes in promoting the cleavage by factor I of C3b to iC3b, C3c, and C3dg. Therefore, a functionally and structurally intact form of soluble CR1 resides in plasma.


Receptors, Complement/analysis , Animals , Centrifugation, Density Gradient , Chromatography, Affinity , Complement C3b Inactivator Proteins/pharmacology , Complement Factor I , Endopeptidases/physiology , Erythrocytes/metabolism , Humans , Lupus Erythematosus, Systemic/blood , Rabbits , Radioimmunoassay/methods , Receptors, Complement/isolation & purification , Receptors, Complement 3b , Solubility
...