Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
World J Surg ; 21(9): 939-42, 1997.
Article in English | MEDLINE | ID: mdl-9361508

ABSTRACT

The major barrier to successful discordant xenogeneic organ transplantation is the phenomenon of hyperacute rejection (HAR). Hyperacute rejection results from the deposition of high-titer preformed antibodies that activate serum complement on the luminal surface of the vascular endothelium, leading to vessel occlusion and graft failure within minutes to hours. Here we describe our strategy to overcome HAR in the pig-to-primate transplant setting, which includes the genetic incorporation into transgenic organs and high level expression of both a novel human bifunctional complement inhibitor and a human blood group enzyme. The expression of the human blood group enzyme is designed to reduce significantly the natural antibody reactivity to the discordant pig tissue, whereas expression of the complement inhibitor results in inhibition of complement-mediated cell activation and lysis. High-level cell surface expression of the complement inhibitor and high-level expression of the human blood group enzyme in vascular endothelium effectively eliminate both the antibody and complement components of the massive inflammatory response to the xenogeneic tissue. Elimination of HAR will establish inroads into understanding the cellular immune response toward the discordant tissue. It is conceivable that standard immunosuppressive regimens routinely practiced with allotransplantation can also be effective drug therapies for xenotransplantation. Therefore it is critical to develop a system that tests these possibilities in order to solve an ever-growing need for donor organs.


Subject(s)
Genetic Engineering/methods , Graft Rejection/prevention & control , Organ Preservation , Organ Transplantation , Transplantation, Heterologous/methods , Animals , Humans , Immunosuppression Therapy/methods
2.
Mol Immunol ; 34(6): 441-52, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9307060

ABSTRACT

Discordant xenografts surviving the initial hyperacute rejection phase may be subject to cellular rejection processes mediated by infiltrating leukocytes including T cells, NK cells and monocytes. The stable adhesion of these cell types to endothelial cells is due to the molecular interaction of the integrins VLA-4 and LFA-1 with their ligands vascular cell adhesion molecule (VCAM) and ICAM-1 present on the endothelial cells. Human VLA-4 binds to porcine VCAM, and blocking mAbs specific for porcine VCAM have been developed. We have localized the epitope of the anti-porcine VCAM blocking mAbs 2A2 and 3F4 to domains 1 and 2, respectively. Humanized antibodies (IgG4 isotype) were constructed from these anti-porcine VCAM antibodies and demonstrated to inhibit adhesion of Ramos, Jurkat and YT cells, as well as purified resting and activated human T cells, to porcine aortic endothelial cells (PAEC). These cell types express both LFA-1 as well as VLA-4, suggesting blockade of human VLA-4 interaction with porcine VCAM may alone be sufficient to significantly impair adhesion of human leukocytes to porcine endothelial cells. The chimeric anti-porcine VCAM (pVCAM) HuG4 antibodies promoted increased adhesion of Fc receptor (FcR) positive cells such as U937 monocytic cells to PAEC. In contrast, chimeric anti-porcine VCAM antibodies created using the CH1 and hinge region from human IgG2 and the CH2 and CH3 regions from human IgG4 (HuG2/G4 antibodies) inhibited binding of FcR positive cells to PAEC. These chimeric anti-pVCAM antibodies should allow delineation of the in vivo role of VLA-4/VCAM interaction in porcine-to-primate xenotransplants. Further, the design of the HuG2/G4 antibodies should render them efficacious in multiple settings requiring elimination of FcR binding.


Subject(s)
Cell Adhesion , Endothelium, Vascular/cytology , Leukocytes/cytology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Binding, Competitive , Cell Line , Epitope Mapping , Humans , Immunoglobulin G/chemistry , Molecular Sequence Data , Receptors, Fc/metabolism , Recombinant Fusion Proteins , Swine , Vascular Cell Adhesion Molecule-1/immunology
3.
Curr Opin Biotechnol ; 7(6): 641-5, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8939635

ABSTRACT

Over the past few years, several major advances have occurred in the understanding of how the humoral and cellular immune system of humans recognizes and destroys transplanted cells, tissues and organs derived from animal sources. Consequently, armed with this new knowledge, several laboratories have now developed novel immunoprotective technologies that may allow xenotransplantation to be clinically feasible.


Subject(s)
Organ Transplantation , Transplantation Immunology , Transplantation, Heterologous , Animals , Cost-Benefit Analysis , Graft Rejection/immunology , Humans , Organ Transplantation/economics , Primates , Swine , Tissue and Organ Procurement , Transplantation, Heterologous/economics
4.
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
5.
J Clin Invest ; 98(7): 1602-12, 1996 Oct 01.
Article in English | MEDLINE | ID: mdl-8833909

ABSTRACT

It has been shown that peripheral T cell tolerance can be induced by systemic antigen administration. We have been interested in using this phenomenon to develop antigen-specific immunotherapies for T cell-mediated autoimmune diseases. In patients with the demyelinating disease multiple sclerosis (MS), multiple potentially autoantigenic epitopes have been identified on the two major proteins of the myelin sheath, myelin basic protein (MBP) and proteolipid protein (PLP). To generate a tolerogenic protein for the therapy of patients with MS, we have produced a protein fusion between the 21.5-kD isoform of MBP (MBP21.5) and a genetically engineered form of PLP (deltaPLP4). In this report, we describe the effects of treatment with this agent (MP4) on clinical disease in a murine model of demyelinating disease, experimental autoimmune encephalomyelitis (EAE). Treatment of SJL/J mice with MP4 after induction of EAE either by active immunization or by adoptive transfer of activated T cells completely prevented subsequent clinical paralysis. Importantly, the administration of MP4 completely suppressed the development of EAE initiated by the cotransfer of both MBP- and PLP-activated T cells. Prevention of clinical disease after the intravenous injection of MP4 was paralleled by the formation of long-lived functional peptide-MHC complexes in vivo, as well as by a significant reduction in both MBP- and PLP-specific T cell proliferative responses. Mice treated with MP4 were resistant to disease when rechallenged with an encephalitogenic PLP peptide emulsified in CFA, indicating that MP4 administration had a prolonged effect in vivo. Administration of MP4 was also found to markedly ameliorate the course of established clinical disease. Finally, MP4 therapy was equally efficacious in mice defective in Fas expression. These results support the conclusion that MP4 protein is highly effective in suppressing disease caused by multiple neuroantigen epitopes in experimentally induced demyelinating disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Recombinant Fusion Proteins/therapeutic use , Vaccination , Vaccines, Synthetic/therapeutic use , Adoptive Transfer , Amino Acid Sequence , Animals , Apoptosis , Female , Histocompatibility Antigens , Immune Tolerance , Lymphocyte Activation , Mice , Mice, Inbred MRL lpr , Molecular Sequence Data , Myelin Basic Protein , Myelin Proteolipid Protein , Peptides , Protein Engineering , T-Lymphocytes/immunology , fas Receptor/biosynthesis
7.
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.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
J Virol ; 69(6): 3889-92, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7745740

ABSTRACT

The herpesvirus saimiri genome encodes a complement control protein homolog (CCPH). Stable mammalian cell transfectants expressing a recombinant transmembrane form of CCPH (mCCPH) or a 5'FLAG epitope-tagged mCCPH (5'FLAGmCCPH) conferred resistance to complement-mediated cell damage by inhibiting the lytic activity of human serum complement. The function of CCPH was further defined by showing that the mCCPH and the 5'FLAGmCCPH transfectants inhibited C3 convertase activity and effectively reduced cell surface deposition of the activated complement component, C3d.


Subject(s)
Complement C3-C5 Convertases/antagonists & inhibitors , Complement System Proteins/metabolism , Herpesvirus 2, Saimiriine/metabolism , Membrane Glycoproteins/physiology , Viral Proteins/physiology , Amino Acid Sequence , Base Sequence , DNA Primers , Humans , Membrane Glycoproteins/genetics , Molecular Sequence Data , Viral Proteins/genetics
19.
In Vitro Cell Dev Biol Anim ; 31(6): 473-81, 1995 Jun.
Article in English | MEDLINE | ID: mdl-8589892

ABSTRACT

We have developed a novel isolation technique for harvesting human capillary endothelial cells. We compared the use of either Ulex Europaeus Agglutinin (UEA) lectin or anti-platelet endothelial cell adhesion molecule (PECAM) antibody conjugated to magnetic beads for the ability to isolate and maintain pure cultures of human capillary endothelial cells. Cells isolated using either method actively scavenged DiI-acetylated-low density lipoprotein and expressed von Willebrand factor (vWf) up to four passages as assessed by immunofluorescent labeling. Endothelial cells isolated using the anti-PECAM antibody method maintained these endothelial-specific properties for up to 12 passages while the percentage of UEA selected cells expressing these properties decreased during increasing passage number. Furthermore, while both techniques yielded cells that bind UEA at Passage six, only the antibody selected cells expressed the normal pattern of endothelial-specific cellular adhesion molecules as assessed by flow cytometry. Both cell isolates were cultured within a three-dimensional matrix of type I collagen, the antibody selected cells formed tubelike structures within 2 days, while the lectin selected cells did not. The antibody selected capillary endothelial cells were transduced with a retroviral vector containing the human growth hormone cDNA and were found to secrete growth hormone from both two- and three-dimensional cultures. We propose that anti-PECAM antibodies linked to a solid support provide a highly selective step in the isolation and maintenance of pure populations of human capillary endothelial cells from abdominal wall liposuction remnants.


Subject(s)
Adipose Tissue/cytology , Antibodies, Monoclonal/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Cell Adhesion Molecules/immunology , Cell Separation/methods , Endothelium, Vascular/cytology , Lectins , Plant Lectins , Abdomen , Animals , Cell Transformation, Viral , Cells, Cultured , Growth Hormone/biosynthesis , Humans , Platelet Endothelial Cell Adhesion Molecule-1 , Vascular Cell Adhesion Molecule-1/biosynthesis , von Willebrand Factor/biosynthesis
20.
Hum Gene Ther ; 6(4): 429-35, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7612700

ABSTRACT

The rapid inactivation of murine-derived retroviral vectors in human or nonhuman primate sera is largely attributed to the activity of complement mediated through the classical pathway. In this study, we have further investigated the relationship between the human complement cascade and retrovirus inactivation. Preincubation in normal human serum effectively inactivated LXSN retroviral vector particles, whereas the vector maintained the ability to transduce cells following incubation in sera deficient in either the C1, C2, C3, C5, C6, C8, or C9 human complement proteins. Preincubation of serum with monoclonal antibodies (mAbs) that functionally block specific complement components, including C5, C6, C8, and C9, successfully protected the LXSN vector from complement-mediated inactivation. Treatment of serum with cobra venom factor, which consumes terminal complement, also effectively protected the vector from inactivation. LXSN vector survival in serum corresponded inversely to the level of complement activity following treatment of serum with anti-C5 mAb as assessed in an erythrocyte hemolytic assay. Additionally, pretreatment of human whole blood with anti-C5 mAb effectively inhibited inactivation of the LXSN vector. Taken together, these data demonstrate that formation of the membrane attack complex (MAC, C5b-9) is required for the inactivation of the murine-based LXSN retroviral vector in human blood and that this process can be abrogated with the use of soluble complement inhibitors.


Subject(s)
Complement Inactivator Proteins/pharmacology , Complement Membrane Attack Complex/antagonists & inhibitors , Genetic Vectors/blood , Moloney murine leukemia virus/immunology , 3T3 Cells , Animals , Antibodies, Monoclonal/pharmacology , Complement C5/immunology , Complement Membrane Attack Complex/immunology , Complement System Proteins/deficiency , Elapid Venoms/pharmacology , Genetic Vectors/immunology , Humans , Mice , Moloney murine leukemia virus/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...