Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Am Heart J ; 141(4): 684-90, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11275938

ABSTRACT

BACKGROUND: Although activation of the complement system in myocardial infarction and cardiopulmonary bypass has been shown to contribute to myocardial injury, its role in congestive heart failure (CHF) is unknown. The purpose of this study was to determine the presence of terminal complement activation and its relation to clinical outcomes in patients with CHF. METHODS: We measured serum levels of the terminal complement complex C5b-9 in 36 patients with symptomatic heart failure and left ventricular ejection fraction <40%. We compared the serum C5b-9 levels of these patients with CHF with a group of 12 age-matched control patients. Combined clinical outcomes (death, urgent heart transplantation, or hospitalization with worsening heart failure) at 6 months were determined. RESULTS: The serum C5b-9 [median (25th to 75th percentiles)] levels in 36 patients with CHF [101.5 ng/mL (40 to 164)] were significantly (P =.003) higher than in the 12 control patients [36.5 ng/mL (22 to 50)]. Significantly more of the patients with CHF with the highest levels of C5b-9 (highest 50th percentile) had New York Heart Association class IV symptoms (67% vs 33%; P =.04) and adverse clinical outcomes by 6 months (56% vs 17%; P =.02) compared with the patients with CHF with lower levels (lowest 50th percentile). CONCLUSIONS: We have described a significant elevation in circulating C5b-9, the terminal complement complex, in patients with symptomatic heart failure and have observed an association between high levels of C5b-9 and near-term adverse events.


Subject(s)
Complement Activation , Complement Membrane Attack Complex/analysis , Heart Failure/blood , Heart Failure/physiopathology , Aged , Female , Hemodynamics , Humans , Male , Middle Aged , Stroke Volume , Tumor Necrosis Factor-alpha/analysis
2.
Transplantation ; 69(8): 1659-67, 2000 Apr 27.
Article in English | MEDLINE | ID: mdl-10836378

ABSTRACT

BACKGROUND: Rapid leukocyte adherence to donor organ vasculature is a hallmark of hyperacute xenograft rejection. However, the molecular interactions required for leukocyte binding to vascular endothelium have not been characterized. METHODS AND RESULTS: Binding assays performed between human neutrophils and porcine aortic endothelial cells (PAEC) after exposure to human complement demonstrated that adhesion was mediated by both surface-bound C3b and C5b-9 activity. C5b-9-dependent adhesion was blocked by neuraminidase treatment of the neutrophils, suggesting that this binding was mediated by porcine P-selectin. Porcine P-selectin was isolated from a PAEC cDNA library. The porcine P-selectin primary sequence contained an open reading frame encoding 646 amino acids with 82% identity to human P-selectin. Recombinant soluble porcine P-selectin specifically bound to human neutrophils and HL-60 cells. Transfection of COS cells with the full-length porcine P-selectin cDNA resulted in surface expression of the protein and markedly increased the binding of human neutrophils to these cells. The binding of both soluble and COS-expressed porcine P-selectin to human neutrophils was blocked by pretreatment of the neutrophils with neuraminidase or the addition of EDTA. Finally, treatment of PAEC with human thrombin or normal human serum but not purified human C5a- or C8-deficient human serum resulted in the rapid expression of porcine P-selectin on the cell surface. CONCLUSIONS: This report establishes that porcine P-selectin supports the binding of human neutrophils to PAEC in vitro. Further, these data suggest that sublytic deposition of C5b-9 during hyperacute rejection results in the expression of porcine P-selectin, which may contribute to the rapid adhesion of neutrophils to porcine xenografts.


Subject(s)
Complement System Proteins/physiology , Endothelium, Vascular/physiology , Leukocytes/physiology , P-Selectin/physiology , Swine/immunology , Amino Acid Sequence/genetics , Animals , Aorta/cytology , Aorta/physiology , COS Cells , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cells, Cultured , Cloning, Molecular , Complement C3b/physiology , Complement Membrane Attack Complex/physiology , Endothelium, Vascular/cytology , Humans , Molecular Sequence Data , Neutrophils/drug effects , Neutrophils/physiology , P-Selectin/genetics , Recombinant Proteins/pharmacology , Transfection
3.
Am J Pathol ; 156(5): 1549-56, 2000 May.
Article in English | MEDLINE | ID: mdl-10793066

ABSTRACT

The complement system plays an important role in mediating tissue injury after oxidative stress. The role of mannose-binding lectin (MBL) and the lectin complement pathway (LCP) in mediating complement activation after endothelial oxidative stress was investigated. iC3b deposition on hypoxic (24 hours; 1% O(2))/reoxygenated (3 hours; 21% O(2)) human endothelial cells was attenuated by N-acetyl-D-glucosamine or D-mannose, but not L-mannose, in a dose-dependent manner. Endothelial iC3b deposition after oxidative stress was also attenuated in MBL-deficient serum. Novel, functionally inhibitory, anti-human MBL monoclonal antibodies attenuated MBL-dependent C3 deposition on mannan-coated plates in a dose-dependent manner. Treatment of human serum with anti-MBL monoclonal antibodies inhibited MBL and C3 deposition after endothelial oxidative stress. Consistent with our in vitro findings, C3 and MBL immunostaining throughout the ischemic area at risk increased during rat myocardial reperfusion in vivo. These data suggest that the LCP mediates complement activation after tissue oxidative stress. Inhibition of MBL may represent a novel therapeutic strategy for ischemia/reperfusion injury and other complement-mediated disease states.


Subject(s)
Complement Activation/physiology , Oxidative Stress , Acetylglucosamine/pharmacology , Animals , Carrier Proteins/immunology , Carrier Proteins/pharmacology , Carrier Proteins/physiology , Cell Line , Collectins , Complement Activation/drug effects , Complement C3b/analysis , Complement C3b/drug effects , Complement C3b/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Hypoxia , Immunohistochemistry , Lectins/physiology , Male , Mannose/pharmacology , Mice , Mice, Inbred BALB C , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Rats , Rats, Inbred Lew
4.
Transplantation ; 68(11): 1643-51, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10609940

ABSTRACT

BACKGROUND: The present study was undertaken to determine whether anti-complement 5 (C5) monoclonal antibodies (mAb) prevent hyperacute rejection (HAR) in a rat-to-presensitized mouse heart transplantation model and whether these mAb, combined with cyclosporine (CsA) and cyclophosphamide (CyP), can achieve long-term graft survival. METHODS: BALB/c mice were presensitized with 2x10(7) splenocytes from Lewis rats 14 days before grafting. Heart grafts from Lewis rats were heterotopically transplanted into BALB/c mice. Presensitized mice were treated with either anti-C5 mAb or a combination of anti-C5 mAb, CsA, and CyP. Controls included: presensitized mice with no treatment, presensitized mice treated with either CsA + CyP or IgG, and nonpresensitized mice with either no treatment or with CsA + CyP treatment. RESULTS: Although typical features of HAR were evident in the presensitized grafts, the mAb completely inhibited complement activation and successfully prevented HAR. Despite complement inactivation, the graft was rejected on postoperative day 6 with acute vascular rejection (AVR) also known as delayed xenograft rejection (DXR). Notably, this type of rejection cannot be effectively overcome by CsA and CyP. CONCLUSIONS: We conclude that (1) anti-C5 mAb prevents HAR, (2) AVR/DXR still occurs when HAR is prevented by complement inactivation, and (3) AVR/DXR cannot be overcome by conventional immunosuppression. These data suggest that anti-C5 mAb may be valuable for preventing HAR in future clinical xenotransplantation and that additional interventions may be required to address AVR/DXR.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Complement C5/immunology , Complement Inactivator Proteins/therapeutic use , Graft Rejection/prevention & control , Heart Transplantation/immunology , Transplantation, Heterologous/immunology , Acute Disease , Animals , Antibodies, Heterophile/analysis , Graft Survival/drug effects , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Myocardium/metabolism , Rats , Rats, Inbred Lew , Treatment Outcome
5.
J Thorac Cardiovasc Surg ; 118(3): 460-6, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10469960

ABSTRACT

OBJECTIVE: Complement activation is induced by cardiopulmonary bypass, and previous work found that late complement components (C5a, C5b-9) contribute to neutrophil and platelet activation during bypass. In the present study, we blocked C5b-9 formation during extracorporeal recirculation of whole blood to assess whether the membrane attack complex was responsible for both platelet and leukocyte activation. METHODS: In a simulated extracorporeal model that activates complement (C3a and sC5b-9), platelets (CD62P expression, leukocyte-platelet conjugate formation), and leukocytes (increased CD11b expression and neutrophil elastase), we examined an anti-human C8 monoclonal antibody that inhibits C5b-9 generation for its effects on cellular activation. RESULTS: Anti-C8 significantly inhibited sC5b-9 formation but did not block C3a generation. Anti-C8 also significantly inhibited the increase in platelet CD62P and monocyte-platelet conjugate formation seen with control circulation. Moreover, compared with control circulation, in which the number of circulating platelets fell by 45%, addition of anti-C8 completely preserved platelet counts. In contrast to blockade of both C5a and sC5b-9 during simulated extracorporeal circulation, neutrophil activation was not inhibited by anti-C8. However, circulating neutrophil and monocyte counts were preserved by addition of anti-C8 to the extracorporeal circuit. CONCLUSIONS: The membrane attack complex, C5b-9, is the major complement determinant of platelet activation during extracorporeal circulation, whereas C5b-9 blockade has little effect on neutrophil activation. These data also suggest a role for platelet activation or C5b-9 (or both) in the loss of monocytes and neutrophils to the extracorporeal circuit.


Subject(s)
Antibodies, Monoclonal/pharmacology , Complement Membrane Attack Complex/physiology , Extracorporeal Circulation , Neutrophil Activation , Platelet Activation , Blood Platelets/drug effects , Blood Platelets/metabolism , Complement Activation , Complement C3a/drug effects , Complement C8/immunology , Complement Membrane Attack Complex/antagonists & inhibitors , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Leukocyte Count , Leukocyte Elastase/metabolism , Macrophage-1 Antigen/metabolism , Neutrophils/enzymology , P-Selectin/metabolism , Platelet Count , Reference Values
6.
Circulation ; 97(22): 2259-67, 1998 Jun 09.
Article in English | MEDLINE | ID: mdl-9631876

ABSTRACT

BACKGROUND: Myocardial ischemia and reperfusion (MI/R)-induced tissue injury involves necrosis and apoptosis. However, the precise contribution of apoptosis to cell death, as well as the mechanism of apoptosis induction, has not been delineated. In this study, we sought to define the contribution of the activated terminal complement components to apoptosis and necrosis in a rat model of MI/R injury. METHODS AND RESULTS: Monoclonal antibodies (mAbs; 18A and 16C) raised against the rat C5 complement component bound to purified rat C5 (ELISA). 18A effectively blocked C5b-9-mediated cell lysis and C5a-induced chemotaxis of rat polymorphonuclear leukocytes (PMNs), whereas 16C had no complement inhibitor activity. A single dose (20 mg/kg i.v.) of 18A blocked >80% of serum hemolytic activity for >4 hours. Administration of 18A before myocardial ischemia (30 minutes) and reperfusion (4 hours) significantly reduced (91%) left ventricular free wall PMN infiltration compared with 16C treatment. Treatment with 18A 1 hour before ischemia or 5 minutes before reperfusion significantly reduced infarct size compared with 16C treatment. A significant reduction in infarct size (42%) was also observed in 18A-treated rats after 30 minutes of ischemia and 7 days of reperfusion. DNA ladders and DNA labeling (eg, TUNEL assay) demonstrated a dramatic reduction in MI/R-induced apoptosis in 18A-treated compared with 16C-treated rats. CONCLUSIONS: Anti-C5 therapy in the setting of MI/R significantly inhibits cell apoptosis, necrosis, and PMN infiltration in the rat despite C3 deposition. We conclude that the terminal complement components C5a and C5b-9 are key mediators of tissue injury in MI/R.


Subject(s)
Apoptosis , Complement System Proteins/physiology , Myocardial Infarction/etiology , Myocardial Ischemia/complications , Myocardial Reperfusion Injury/complications , Animals , Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Complement C5/immunology , Complement Inactivator Proteins/therapeutic use , Creatine Kinase/metabolism , Male , Myocardial Infarction/pathology , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Myocardium/enzymology , Myocardium/pathology , Peroxidase/metabolism , Rats , Rats, Inbred Lew
7.
Mol Immunol ; 33(17-18): 1389-401, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9171898

ABSTRACT

Activation of the complement system contributes significantly to the pathogenesis of numerous acute and chronic diseases. Recently, a monoclonal antibody (5G1.1) that recognizes the human complement protein C5, has been shown to effectively block C5 cleavage, thereby preventing the generation of the pro-inflammatory complement components C5a and C5b-9. Humanized 5G1.1 antibody, Fab and scFv molecules have been produced by grafting the complementarity determining regions of 5G1.1 on to human framework regions. Competitive ELISA analysis indicated that no framework changes were required in the humanized variable regions for retention of high affinity binding to C5, even at framework positions predicted by computer modeling to influence CDR canonical structure. The humanized Fab and scFv molecules blocked complement-mediated lysis of chicken erythrocytes and porcine aortic endothelial cells in a dose-dependent fashion, with complete complement inhibition occurring at a three-fold molar excess, relative to the human C5 concentration. In contrast to a previously characterized anti-C5 scFv molecule, the humanized h5G1.1 scFv also effectively blocked C5a generation. Finally, an intact humanized h5G1.1 antibody blocked human complement lytic activity at concentrations identical to the original murine monoclonal antibody. These results demonstrate that humanized h5G1.1 and its recombinant derivatives retain both the affinity and blocking functions of the murine 5G1.1 antibody, and suggest that these molecules may serve as potent inhibitors of complement-mediated pathology in human inflammatory diseases.


Subject(s)
Complement C5/immunology , Complement Inactivator Proteins/pharmacology , Immunoglobulin Fragments/pharmacology , Immunoglobulin Variable Region/pharmacology , Recombinant Fusion Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Blocking/genetics , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Aorta , Base Sequence , Binding, Competitive/immunology , Chickens , Complement C5a/immunology , Complement Membrane Attack Complex/antagonists & inhibitors , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Enzyme-Linked Immunosorbent Assay , Erythrocytes/immunology , Humans , Immunoglobulin Variable Region/genetics , Molecular Sequence Data , Recombinant Fusion Proteins/pharmacology , Swine
8.
Proc Natl Acad Sci U S A ; 93(16): 8563-8, 1996 Aug 06.
Article in English | MEDLINE | ID: mdl-8710910

ABSTRACT

New Zealand black x New Zealand white (NZB/W) F1 mice spontaneously develop an autoimmune syndrome with notable similarities to human systemic lupus erythematosus. Female NZB/WF1 mice produce high titers of antinuclear antibodies and invariably succumb to severe glomerulonephritis by 12 months of age. Although the development of the immune-complex nephritis is accompanied by abundant local and systemic complement activation, the role of proinflammatory complement components in disease progression has not been established. In this study we have examined the contribution of activated terminal complement proteins to the pathogenesis of the lupus-like autoimmune disease. Female NZB/W F1 mice were treated with a monoclonal antibody (mAb) specific for the C5 component of complement that blocks the cleavage of C5 and thus prevents the generation of the potent proinflammatory factors C5a and C5b-9. Continuous therapy with anti-C5 mAb for 6 months resulted in significant amelioration of the course of glomerulonephritis and in markedly increased survival. These findings demonstrate an important role for the terminal complement cascade in the progression of renal disease in NZB/W F1 mice, and suggest that mAb-mediated C5 inhibition may be a useful approach to the therapy of immune-complex glomerulonephritis in humans.


Subject(s)
Antibodies, Blocking/therapeutic use , Antibodies, Monoclonal/therapeutic use , Complement C5/immunology , Lupus Erythematosus, Systemic/therapy , Animals , Antigen-Antibody Complex , Arthus Reaction , Complement Activation , Complement C5/antagonists & inhibitors , Female , Glomerulonephritis/immunology , Glomerulonephritis/therapy , Heterozygote , Immune Complex Diseases/immunology , Immune Complex Diseases/therapy , Immunotherapy , Kidney/pathology , Mice , Mice, Inbred NZB
9.
Transplantation ; 61(10): 1513-21, 1996 May 27.
Article in English | MEDLINE | ID: mdl-8633381

ABSTRACT

The serious shortage of available donor organs for patients with end stage organ failure who are in need of solid organ transplantation has led to a heightened interest in xenotransplantation. The major barrier to successful discordant xenotransplantation is hyperacute rejection. Hyperacute rejection results from the deposition of preformed antibodies that activate complement on the luminal surface of the vascular endothelium, leading to vessel occlusion and graft failure within minutes to hours. Endogenous membrane-associated complement inhibitors normally protect endothelial cells from autologous complement -- however, these molecules are species-restricted and therefore are ineffective at inhibiting activated xenogeneic complement. To address the pathogenesis of hyperacute rejection in the pig-to-human combination, F1 offspring were generated from a transgenic founder animal that was engineered to express the human terminal complement inhibitor hCD59. High-level cell surface expression of hCD59 was detected in the hearts and kidneys of these transgenic F1 animals, similar to expression levels in human kidney tissue. The hCD59 was expressed on both large vessel and capillary endothelium. Ex vivo perfusion experiments, using human blood as the perfusate, were performed with transgenic porcine hearts and kidneys to evaluate the ability of hCD59 to inhibit hyperacute rejection. These experiments demonstrated that transgenic organs expressing hCD69 resisted hyperacute rejection, as measured by increased organ function for both the hearts and the kidneys, as compared with control pig organs. Hearts from hCD59-expressing animals demonstrated a five-fold prolongation in function compared with controls, 109.8 +/- 20.7 min versus 21.2 +/- 2.9 min (P = 0.164). The hCD59-expressing kidneys also demonstrated significantly prolonged function at 157.8 +/- 27.0 min compared with 60.0 +/- 6.1 min for controls (P = 0.0174). Deposition of C9 neoantigen In the vasculature of porcine organs perfused with human blood was markedly reduced in organs expressing hCD59. These studies demonstrate that C5b-9 plays an important role in hyperacute rejection of a porcine organ perfused with human blood and suggest that donor pigs transgenic for hCD59 may be an integral component of successful clinical xenotransplantation.


Subject(s)
CD59 Antigens/genetics , Graft Rejection , Transplantation, Heterologous/methods , Animals , Animals, Genetically Modified , Base Sequence , CD59 Antigens/metabolism , Complement Activation , Complement C3/metabolism , Complement C9/metabolism , DNA Primers/chemistry , Disease Models, Animal , Heart/physiology , Hemolysis , Humans , Kidney/physiology , Leukocytes, Mononuclear/metabolism , Molecular Sequence Data , Perfusion
10.
Hum Gene Ther ; 7(5): 619-26, 1996 Mar 20.
Article in English | MEDLINE | ID: mdl-8845387

ABSTRACT

The introduction of retroviral vector producer cells (VPC) into tumors as a means of increasing transduction efficiency has recently been employed in human gene therapy trials. However, the fate of these xenogeneic cells in humans is not well understood. In the present study, we used an in vitro model to examine the survival of commonly used VPC lines in serum from humans and various other species. VPC derived from the murine NIH-3T3 cell line, including PA317, Psi CRIP, and GP + E-86, were effectively killed in sera from Old World primates, including human and baboon. Conversely, the same murine cell lines survived exposure to sera from dog, rabbit, rat, and mouse. This pattern of serum killing parallels the occurrence of the anti-alpha-galactosyl natural antibody (Ab) found exclusively in Old World primates. The anti-alpha-galactosyl Ab targets the terminal glycosidic structure Gal alpha 1-3Gal beta 1-4GlcNAc-R (alpha-galactosyl epitope) found on the surface of mammalian cells, excluding Old World primates. All murine-derived VPC tested expressed high levels of the alpha-galactosyl epitope as determined by FACS analysis. VPC killing was complement-mediated, because preincubation of human serum with a functionally blocking anti-C5 mAb completely abolished cell lysis. Furthermore, addition of soluble galactose(alpha 1-3)galactose (Gal alpha 1-3Gal) to human serum or down-regulation of the alpha-galactosyl epitope on the surface of VPC effectively reduced VPC killing, indicating that complement activation by these cells is primarily initiated by natural antibody recognition of the alpha-galactosyl epitope. Finally, VPC incubated with human serum for 8 hr in the presence of complement inhibition continued to produce viable retroviral particles, thus demonstrating a correlation between VPC and particle survival. Taken together, these data suggest that elimination of the alpha-galactosyl epitope or complement blockade may provide a strategy to prolong the survival of VPC and the particles that they produce in vivo.


Subject(s)
Antibodies/immunology , Cell Survival/genetics , Complement System Proteins/immunology , Galactose/immunology , Genetic Vectors , Retroviridae/genetics , Animals , Carbohydrate Sequence , Cells, Cultured , Dogs , Epitopes/immunology , Flow Cytometry , Humans , Mice , Molecular Sequence Data , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Papio/metabolism , Rabbits , Rats , Retroviridae/metabolism
11.
Transplantation ; 60(11): 1194-202, 1995 Dec 15.
Article in English | MEDLINE | ID: mdl-8525509

ABSTRACT

Prevention of hyperacute xenograft rejection in the pig-to-primate combination has been accomplished by removal of natural antibodies, complement depletion with cobra venom factor, or prevention of C3 activation with the soluble complement inhibitor sCR1. Although these strategies effectively prevent hyperacute rejection, they do not address the relative contribution of early (C3a, C3b) versus late (C5a, C5b-9) activated complement components to xenogeneic organ damage. To better understand the role of the terminal complement components (C5a, C5b-9) in hyperacute rejection, an anti-human C5 mAb was developed and tested in an ex vivo model of cardiac xenograft rejection. In vitro studies demonstrated that the anti-C5 mAb effectively blocked C5 cleavage in a dose-dependent manner that resulted in complete inhibition of both C5a and C5b-9 generation. Addition of anti-C5 mAb to human blood used to perfuse a porcine heart prolonged normal sinus cardiac rhythm from a mean time of 25.2 min in hearts perfused with unmodified blood to 79,296, or > 360 min when anti-C5 mAb was added to the blood at 50 micrograms/ml, 100 micrograms/ml, or 200 micrograms/ml, respectively. In these experiments, activation of the classical complement pathway was completely inhibited. Hearts perfused with blood containing the highest concentration of anti-C5 mAb had no histologic evidence of hyperacute rejection and no deposition of C5b-9. These experiments suggest that the activated terminal complement components C5a and C5b-9, but not C3a or C3b, play a major role in tissue damage in this porcine-to-human model of hyperacute rejection. They also suggest that targeted inhibition of terminal complement activation by anti-C5 mAbs may be useful in clinical xenotransplantation.


Subject(s)
Antibodies, Monoclonal/immunology , Complement C5/physiology , Graft Rejection , Heart Transplantation/immunology , Myocardium/immunology , Acute Disease , Animals , Antibodies, Monoclonal/therapeutic use , Complement Activation , Endothelium, Vascular/immunology , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Mice , Mice, Inbred BALB C , Perfusion , Swine , Transplantation, Heterologous
12.
Transplantation ; 60(11): 1284-92, 1995 Dec 15.
Article in English | MEDLINE | ID: mdl-8525523

ABSTRACT

The hyperacute rejection (HAR) of xenotransplanted organs is initiated by the deposition of natural antibodies on donor endothelium followed by the activation of the recipient complement system, which rapidly destroys the graft. Studies of the role of activated complement in HAR have suggested that natural antibody as well as early (C3a, C3b) and late (C5a, C5b-9) activated complement components may contribute to cell activation and damage. Attenuation of HAR has been achieved by blockade of C3 activation with soluble CR1 or consumptive depletion of complement with cobra venom factor; however, similar studies using specific inhibitors of terminal complement components have not been described. To address the contribution of C5a and the membrane attack complex (C5b-9, MAC) to complement-mediated xenogeneic cell and organ damage, we utilized functionally blocking monoclonal antibodies directed against the human terminal complement components C5 and C8. Our data show that both anti-C5 and anti-C8 mAbs protect porcine aortic endothelial cells from membrane damage mediated by human C5b-9. Additionally, both the anti-C5 and anti-C8 mAbs blocked complement-mediated generation of membrane prothrombinase activity on porcine aortic endothelial cells challenged with human serum. To test the ability of these antibodies to attenuate antibody and complement-mediated damage of xenogeneic organs, an ex vivo model was developed wherein isolated rat hearts were perfused with human serum in the presence or absence of the anti-C5 and anti-C8 mAbs. Our data demonstrate that mAbs directed against human C5 and C8 prevented organ damage by human serum complement and suggest that these molecules may serve as potent inhibitors of HAR.


Subject(s)
Complement C5/immunology , Complement C8/immunology , Complement Membrane Attack Complex/immunology , Graft Rejection , Animals , Antibodies, Monoclonal , Complement Activation , Complement Membrane Attack Complex/metabolism , Endothelium, Vascular/immunology , Humans , Myocardium/immunology , Perfusion , Rats , Swine , Thromboplastin/metabolism
13.
Nat Med ; 1(12): 1261-7, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7489406

ABSTRACT

The major obstacle to successful discordant xenotransplantation is the phenomenon of hyperacute rejection (HAR). In the pig-to-primate discordant transplant setting, HAR results from the deposition of high-titre anti-alpha-galactosyl antibodies and complement activation leading to endothelial cell destruction and rapid graft failure. To overcome HAR, we developed an enzymatic carbohydrate remodelling strategy designed to replace expression of the Gal alpha-1,3-Gal xenoepitope on the surface of porcine cells with the non-antigenic universal donor human blood group O antigen, the alpha-1,2-fucosyl lactosamine moiety (H-epitope). Xenogenic cells expressing the human alpha-1,2-fucosyltransferase expressed high levels of the H-epitope and significantly reduced Gal alpha-1,3-Gal expression. As a result, these cells were shown to be resistant to human natural antibody binding and complement-mediated cytolysis.


Subject(s)
Disaccharides/metabolism , Fucosyltransferases/metabolism , RNA, Messenger/metabolism , Animals , Base Sequence , Binding, Competitive , Cell Line , Complement Activation , DNA Primers , Fucosyltransferases/genetics , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Graft Rejection/immunology , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Transfection , Transplantation, Heterologous/immunology , Tumor Cells, Cultured , Galactoside 2-alpha-L-fucosyltransferase
14.
Mol Immunol ; 32(16): 1183-95, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8559143

ABSTRACT

Complement activation has been implicated in the pathogenesis of several human diseases. Recently, a monoclonal antibody, (N19-8) that recognizes the human complement protein C5 has been shown to effectively block the cleavage of C5 into C5a and C5b, thereby blocking terminal complement activation. In this study, a recombinant N19-8 scFv antibody fragment was constructed from the N19-8 variable regions, and produced in both mammalian and bacterial cells. The N19-8 scFv bound human C5 and was as potent as the N19-8 monoclonal antibody at inhibiting human C5b-9-mediated hemolysis of chicken erythrocytes. In contrast, the N19-8 scFv only partially retained the ability of the N19-8 monoclonal antibody to inhibit C5a generation. To investigate the ability of the N19-8 scFv to inhibit complement-mediated tissue damage, complement-dependent myocardial injury was induced in isolated mouse hearts by perfusion with Krebs-Henseleit buffer containing 6% human plasma. The perfused hearts sustained extensive deposition of human C3 and C5b-9, resulting in increased coronary artery perfusion pressure, end-diastolic pressure, and a decrease in heart rate until the hearts ceased beating approximately 10 min after addition of plasma. Hearts treated with human plasma supplemented with either the N19-8 monoclonal antibody or the N19-8 scFv did not show any detectable changes in cardiac performance for at least 1 hr following the addition of plasma. Hearts treated with human plasma alone showed extensive deposition of C3 and C5b-9, while hearts treated with human plasma containing N19-8 scFv showed extensive deposition of C3, but no detectable deposition of C5b-9. Administration of a 100 mg bolus dose of N19-8 scFv to rhesus monkeys inhibited the serum hemolytic activity by at least 50% for up to 2 hr. Pharmacokinetic analysis of N19-8 scFv serum levels suggested a two-compartment model with a T1/2 alpha of 27 min. Together, these data suggest the recombinant N19-8 scFv is a potent inhibitor of the terminal complement cascade and may have potential in vivo applications where short duration inhibition of terminal complement activity is desirable.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Complement C5/immunology , Immunoglobulin Variable Region/immunology , Myocardium/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Base Sequence , Blood Pressure/drug effects , Complement Activation/drug effects , Complement C5/metabolism , Heart Rate/drug effects , Hemolysis/drug effects , Humans , Immunoglobulin Variable Region/chemistry , Macaca mulatta , Mice , Molecular Sequence Data , Myocardium/pathology , Perfusion , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
15.
J Immunol ; 155(9): 4135-8, 1995 Nov 01.
Article in English | MEDLINE | ID: mdl-7594566

ABSTRACT

Human cells express cell surface complement regulatory molecules that inhibit the activity of the C3/C5 convertases (DAF, MCP, CR1) or inhibit the membrane attack complex (CD59). A single molecule that inhibits both the convertase activity and formation of the membrane attack complex has never been characterized. To this end, we have developed two reciprocal chimeric complement inhibitors (CD, NH2-CD59-DAF-GPI; and DC, NH2-DAF-CD59-GPI) that contain the functional domains of decay accelerating factor (DAF; CD55) and CD59. Cell surface expression of the CD and DC chimeric proteins was detected with DAF- and CD59-specific antisera. Cell surface C3d deposition was inhibited on cells expressing the chimeric molecules, thereby indicating that the DAF moiety was functional in both molecules. Conversely, Ab-blocking experiments demonstrated that only the DC molecule retained CD59 function. Therefore, the DC molecule represents a novel potent chimeric bifunctional complement inhibitor that retains the functional domains of two distinct complement regulatory molecules.


Subject(s)
CD55 Antigens/physiology , CD59 Antigens/physiology , Complement C3-C5 Convertases/antagonists & inhibitors , Complement Inactivator Proteins/physiology , Complement Membrane Attack Complex/biosynthesis , Recombinant Fusion Proteins/physiology , Animals , Base Sequence , CD55 Antigens/genetics , CD59 Antigens/genetics , Cell Line , Complement C3-C5 Convertases/genetics , Complement Inactivator Proteins/biosynthesis , Complement Inactivator Proteins/genetics , Complement Membrane Attack Complex/antagonists & inhibitors , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Engineering , Recombinant Fusion Proteins/biosynthesis
16.
J Exp Med ; 182(5): 1345-55, 1995 Nov 01.
Article in English | MEDLINE | ID: mdl-7595205

ABSTRACT

Type C retroviruses endogenous to various nonprimate species can infect human cells in vitro, yet the transmission of these viruses to humans is restricted. This has been attributed to direct binding of the complement component C1q to the viral envelope protein p15E, which leads to classical pathway-mediated virolysis in human serum. Here we report a novel mechanism of complement-mediated type C retrovirus inactivation that is initiated by the binding of "natural antibody" [Ab] (anti-alpha-galactosyl Ab) to the carbohydrate epitope Gal alpha 1-3Gal beta 1-4GlcNAc-R expressed on the retroviral envelope. Complement-mediated inactivation of amphotropic retroviral particles was found to be restricted to human and other Old World primate sera, which parallels the presence of anti-alpha-galactosyl natural Ab. Blockade or depletion of anti-alpha-galactosyl Ab in human serum prevented inactivation of both amphotropic and ecotropic murine retroviruses. Similarly, retrovirus was not killed by New World primate serum except in the presence of exogenous anti-alpha-galactosyl Ab. Enzyme-linked immunosorbent assays revealed that the alpha-galactosyl epitope was expressed on the surface of amphotropic and ecotropic retroviruses, and Western blot analysis further localized this epitope to the retroviral envelope glycoprotein gp70. Finally, down-regulation of this epitope on the surface of murine retroviral particle producer cells rendered them, as well as the particles liberated from these cells, resistant to inactivation by human serum complement. Our data suggest that anti-alpha-galactosyl Ab may provide a barrier for the horizontal transmission of retrovirus from species that express the alpha-galactosyl epitope to humans and to other Old World primates. Further, these data provide a mechanism for the generation of complement-resistant retroviral vectors for in vivo gene therapy applications where exposure to human complement is unavoidable.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Blood/virology , Cebidae/immunology , Cercopithecidae/immunology , Epitopes/immunology , Galactose/immunology , Leukemia Virus, Murine/physiology , Retroviridae Proteins, Oncogenic/immunology , Viral Envelope Proteins/immunology , 3T3 Cells , Animals , Antibodies, Viral/immunology , Antigens, Viral/biosynthesis , Blood/immunology , Carbohydrate Sequence , Cebidae/blood , Cercopithecidae/blood , Complement System Proteins/immunology , Humans , Immunity, Innate , Mammals/blood , Mammals/immunology , Mice , Molecular Sequence Data , Moloney murine leukemia virus/immunology , Retroviridae Proteins, Oncogenic/biosynthesis , Species Specificity , Viral Envelope Proteins/biosynthesis
17.
Proc Natl Acad Sci U S A ; 92(19): 8955-9, 1995 Sep 12.
Article in English | MEDLINE | ID: mdl-7568051

ABSTRACT

Activated components of the complement system are potent mediators of inflammation that may play an important role in numerous disease states. For example, they have been implicated in the pathogenesis of inflammatory joint diseases including rheumatoid arthritis (RA). To target complement activation in immune-mediated joint inflammation, we have utilized monoclonal antibodies (mAbs) that inhibit the complement cascade at C5, blocking the generation of the major chemotactic and proinflammatory factors C5a and C5b-9. In this study, we demonstrate the efficacy of a mAb specific for murine C5 in the treatment of collagen-induced arthritis, an animal model for RA. We show that systemic administration of the anti-C5 mAb effectively inhibits terminal complement activation in vivo and prevents the onset of arthritis in immunized animals. Most important, anti-C5 mAb treatment is also highly effective in ameliorating established disease. These results demonstrate a critical role for activated terminal complement components not only in the induction but also in the progression of collagen-induced arthritis and suggest that C5 may be an attractive therapeutic target in RA.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/drug therapy , Complement C5/immunology , Immunization, Passive , Synovitis/drug therapy , Animals , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/prevention & control , Collagen/pharmacology , Disease Models, Animal , Disease Progression , Extremities/pathology , Joints/pathology , Male , Mice , Mice, Inbred DBA , Synovitis/chemically induced , Synovitis/prevention & control
18.
J Clin Invest ; 96(3): 1564-72, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7657827

ABSTRACT

Complement activation contributes to the systemic inflammatory response induced by cardiopulmonary bypass. At the cellular level, cardiopulmonary bypass activates leukocytes and platelets; however the contribution of early (3a) versus late (C5a, soluble C5b-9) complement components to this activation is unclear. We used a model of simulated extracorporeal circulation that activates complement (C3a, C5a, and C5b-9 formation), platelets (increased percentages of P-selectin-positive platelets and leukocyte-platelet conjugates), and neutrophils (upregulated CD11b expression). to specifically target complement activation in this model, we added a blocking mAb directed at the human C5 complement component and assessed its effect on complement and cellular activation. Compared with a control mAB, the anti-human C5 mAb profoundly inhibited C5a and soluble C5b-9 generation and serum complement hemolytic activity but had no effect on C3a generation. Additionally, the anti-human C5 mAb significantly inhibited neutrophil CD11b upregulation and abolished the increase in P-selectin-positive platelets and leukocyte-platelet conjugate formation compared to experiments performed with the control mAb. This suggests that the terminal components C5a and C5b-9, but not C3a, directly contribute to platelet and neutrophil activation during extracorporeal circulation. Furthermore, these data identify the C5 component as a site for therapeutic intervention in cardiopulmonary bypass.


Subject(s)
Antibodies, Monoclonal/pharmacology , Blood Platelets/physiology , Complement Activation , Complement C5a/physiology , Complement Membrane Attack Complex/physiology , Extracorporeal Circulation , Hemolysis , Leukocytes/physiology , Platelet Activation , CD11 Antigens/blood , Cardiopulmonary Bypass , Complement C5a/antagonists & inhibitors , Complement C5a/immunology , Complement Membrane Attack Complex/antagonists & inhibitors , Complement Membrane Attack Complex/immunology , Humans , Kinetics , Models, Biological , Neutrophils/immunology , Neutrophils/physiology , Reference Values , Time Factors
20.
J Immunol Methods ; 184(1): 123-38, 1995 Jul 17.
Article in English | MEDLINE | ID: mdl-7622864

ABSTRACT

Inhibition of complement system activation requires the development of soluble nonimmunogenic inhibitors with good tissue penetrating abilities that are themselves unable to activate complement. Chimeric mouse/human Fabs capable of blocking the activity of complement proteins are likely to fulfill these criteria. Several monoclonal antibodies that inhibit the activation of the human complement system have recently been developed. To examine the properties of chimeric Fab derived from these monoclonal antibodies, we have developed an expression system which allows the rapid production of milligram quantities of chimeric Fab. Both the chimeric light chain and the chimeric Fd were co-expressed from the same vector, pAPEX-3P. This vector contains the SV40 origin of replication, which allows the rapid production of chimeric Fab in COS cells for preliminary characterization. Additionally, pAPEX-3P contains the Epstein-Barr virus origin of replication and a puromycin selectable marker for maintenance as a stable episome in human cell lines. A production system consisting of transfected 293-EBNA cells cultured in serum free medium followed by protein G-Sepharose chromatography of the conditioned medium was found to be sufficient for the rapid production of purified chimeric Fab. Here we have utilized this expression system to demonstrate that an anti-human C5 chimeric Fab was a potent inhibitor of complement activation in both in vitro activation assays and an ex vivo model of complement-mediated tissue damage.


Subject(s)
Complement C5/immunology , Genetic Vectors , Immunoglobulin Fab Fragments/genetics , Animals , Antibodies, Monoclonal/immunology , Base Sequence , Cell Line , Chickens , Cloning, Molecular , DNA Primers , Genetic Vectors/genetics , Herpesvirus 4, Human/genetics , Humans , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/immunology , Mice , Molecular Sequence Data , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Simian virus 40/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...