Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Transplant Proc ; 40(2): 554-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374127

ABSTRACT

Cartilage engineering is the object of intense research as a result of major medical needs and therapeutic prospects. Porcine xenogeneic cells/tissues may help in the development of clinical applications such as articular cartilage repair. However, unmodified porcine cartilage is rejected in primates by humoral and cellular mechanisms. We previously showed that porcine articular chondrocytes (PAC) isolated from H-transferase (HT) transgenic pigs show markedly reduced expression of the Galalpha1,3Gal antigen (alphaGal) and prolonged survival when transplanted into alpha1,3galactosyltransferase-deficient mice. In this work, we further studied the protective mechanisms of HT transgenic expression in cartilage, particularly its effects on monocyte adhesion. To this end, PAC isolated from control and HT transgenic pigs were assayed for human complement deposition and adhesion to the human monoblastic cell line U937. Consistent with a reduction in complement activation by the classical pathway, the HT transgenic PAC showed a 2-fold reduction in the deposition of complement components C4 and C3 relative to controls. Adhesion of U937 cells to HT PAC was also diminished under various conditions. This reduction was more dramatic at high effector:target ratios and especially observed when combined with anti-alphaGal antibodies (5-fold difference). Nevertheless, this effect was also observed in the absence of anti-alphaGal. antibodies and after tumor necrosis factor treatment. These results suggest that HT expression on porcine chondrocytes protects them from both humoral and cellular rejection.


Subject(s)
Cartilage/physiology , Cell Adhesion/physiology , Fucosyltransferases/genetics , Swine/genetics , Animals , Animals, Genetically Modified , Antibody Formation , Cartilage/enzymology , Cartilage/immunology , Complement System Proteins/physiology , Humans , Immunity, Cellular , Transplantation, Heterologous/physiology , U937 Cells , Galactoside 2-alpha-L-fucosyltransferase
2.
Transplant Proc ; 39(7): 2443-6, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17889214

ABSTRACT

Clinical solid organ xenotransplantation is precluded by the strong immune response that results in rejection of pig xenografts in primate models. Innate immunity seems to play a major role in this process. In particular, tumor necrosis factor (TNF), produced by natural killer cells and macrophages, contributes to xenograft rejection by promoting endothelial cell activation and the recruitment of inflammatory cells. To further elucidate its molecular mechanism, we cloned the full-length cDNA of porcine TNF-Receptor 2 (pTNFR2, p75) by reverse transcriptase polymerase chain reaction (PCR) of total RNA isolated from porcine peripheral blood mononuclear cells. To this end, we used degenerate primers based on the sequences of the mouse, rat, and human homologues. Two PCR fragments were obtained that contained the pTNFR2 sequence, but differed in size. The shorter clones lacked the sequence corresponding to exon 4 by homology but identical for the rest, suggesting there is an alternative spliced mRNA variant of the porcine receptor. The predicted protein sequence (461 amino acids, containing exon 4) exhibited 72.5% identity to the human TNFR2 and 58.7% to the mouse molecule. By predicted protein sequence analysis, we determined that it comprised the four TNFR cysteine-rich repeats conserved between species. However, the molecule missing exon 4 lacks one cysteine-rich repeat. To assess function, we produced two recombinant proteins containing the extracellular domain of each pTNFR2 variant fused to the Fc portion of human IgG1. Next, we examined their ability to inhibit human TNF-mediated activation of porcine aortic endothelial cells. The addition of the whole pTNFR2 fusion protein to the TNF treatment blocked the up-regulation of activation markers. However, the fusion protein lacking exon 4 failed to effectively counteract TNF effects. These two pTNFR2 isoforms may play differential roles in the process of xenograft rejection.


Subject(s)
Receptors, Tumor Necrosis Factor, Type II/physiology , Transplantation, Heterologous/physiology , Animals , DNA Primers , DNA, Complementary/genetics , Exons , Graft Rejection/genetics , Immunity, Innate , Mice , Primates , Protein Isoforms/genetics , RNA/blood , RNA/genetics , RNA/isolation & purification , Receptors, Tumor Necrosis Factor, Type I/physiology , Receptors, Tumor Necrosis Factor, Type II/genetics , Reverse Transcriptase Polymerase Chain Reaction , Swine , Transplantation, Heterologous/immunology
3.
Xenotransplantation ; 11(6): 491-502, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15479458

ABSTRACT

BACKGROUND: Delayed rejection of xenografts is a major hurdle that needs to be addressed to achieve long-term engraftment in the pig-to-primate transplant setting. Both vascular and avascular xenografts are susceptible to a delayed rejection process that comprises humoral and cellular responses. Tumor necrosis factor (TNF) is believed to play a role in this process by promoting cell activation, apoptosis and the recruitment of inflammatory cells. To address this problem, we engineered the donor cell in such a way that it could block both human and porcine TNF. METHODS: We produced a recombinant fusion protein containing the extracellular domain of the porcine TNF-Receptor 1 and an IgG Fc moiety (pTNFR1Ig). We first evaluated by flow cytometry the pTNFR1Ig capacity to prevent TNF alpha-induced expression of SLAI, SLAII, VCAM-1, ICAM-1 and E-selectin on the cell surface of porcine aortic endothelial cells (PAEC). The effect on TNF alpha-mediated cell death was also assessed by propidium iodide staining after incubating PAEC with TNF alpha plus cycloheximide for 24 h. PAEC and porcine fibroblasts were subsequently engineered by retroviral infection to express and secrete pTNFR1Ig and their resistance to the TNF alpha effects was tested in vitro. Finally, we transplanted mock-control and pTNFR1Ig-expressing PAEC under the kidney capsule of BALB/c mice in the absence of immunosuppression and examined the degree of rejection at 2 and 3 weeks post-transplantation. RESULTS: Treatment with pTNFR1Ig resulted in a very potent blockade of human, porcine and murine TNF alpha activity on porcine cells. It inhibited the upregulation of all cell surface markers of activation tested as well as the TNF alpha-mediated cell death. Moreover, pTNFR1Ig-expressing PAEC showed prolonged engraftment in a pig-to-mouse xenotransplant model. CONCLUSIONS: Incorporation of strategies that block TNF may prove useful in the development of xenografts resistant to delayed rejection.


Subject(s)
Graft Rejection/prevention & control , Graft Survival/drug effects , Immunoglobulin Heavy Chains/pharmacology , Kidney Transplantation , Recombinant Fusion Proteins/pharmacology , Transplantation, Heterologous , Animals , Cell Line , Flow Cytometry , Gene Expression , Graft Rejection/drug therapy , Humans , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin gamma-Chains , Mice , Mice, Inbred BALB C , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Fusion Proteins/metabolism , Swine , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
4.
Nat Biotechnol ; 18(9): 949-53, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973214

ABSTRACT

Here we describe transplantation of olfactory ensheathing cells (OECs) or Schwann cells derived from transgenic pigs expressing the human complement inhibitory protein, CD59 (hCD59), into transected dorsal column lesions of the spinal cord of the immunosuppressed rat to induce axonal regeneration. Non-transplanted lesion-controlled rats exhibited no impulse conduction across the transection site, whereas in animals receiving transgenic pig OECs or Schwann cells impulse conduction was restored across and beyond the lesion site for more than a centimeter. Cell labeling indicated that the donor cells migrated into the denervated host tract. Conduction velocity measurements showed that the regenerated axons conducted impulses faster than normal axons. By morphological analysis, the axons seemed thickly myelinated with a peripheral pattern of myelin expected from the donor cell type. These results indicate that xenotranplantation of myelin-forming cells from pigs genetically altered to reduce the hyperacute response in humans are able to induce elongative axonal regeneration and remyelination and restore impulse conduction across the transected spinal cord.


Subject(s)
Axons/physiology , CD59 Antigens/genetics , Olfactory Nerve/cytology , Regeneration , Spinal Cord/physiology , Transplantation, Heterologous/methods , Animals , Animals, Genetically Modified , Axons/ultrastructure , CD59 Antigens/metabolism , Cell Separation , Electrophysiology , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Immunosuppression Therapy , Models, Biological , Olfactory Nerve/metabolism , Rats , Rats, Wistar , Schwann Cells/metabolism , Sciatic Nerve/metabolism , Spinal Cord/ultrastructure , Swine , Transgenes
5.
J Immunol ; 164(12): 6340-8, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10843688

ABSTRACT

Recent studies have suggested that soluble forms of B7-1 and B7-2 may exist, but transcripts that code for these molecules have not been previously described. In this study, we report the cloning and characterization of an alternatively spliced soluble form of porcine B7-1 (sB7-1) that lacks exons coding for both the transmembrane and cytoplasmic domains. Northern blot analysis of RNA from alveolar macrophages revealed an approximate 3:1 ratio of the transmembrane form of B7-1 mRNA relative to sB7-1 mRNA. Porcine B7-1 was present on the surface of both B and T cells following stimulation with PMA/ionomycin. A histidine-tagged form of porcine sB7-1 (sB7-1-His) interacted with both CD28 and CTLA-4, and effectively blocked IL-2 production from human responder cells stimulated with PHA and either porcine or human stimulator cells. In addition, sB7-1-His inhibited human T cell proliferation in response to porcine or human peripheral blood leukocytes. This study is the first report of an alternatively spliced form of B7 that codes for a soluble protein. Furthermore, these data demonstrate that porcine B7-1 interacts with the human receptors CD28 and CTLA-4, suggesting a potential role for this molecule in pig to human xenotransplantation. Possible physiological functions for the soluble form of B7-1 are discussed.


Subject(s)
Alternative Splicing/immunology , B7-1 Antigen/chemistry , B7-1 Antigen/physiology , Immunoconjugates , Abatacept , Adult , Amino Acid Sequence , Animals , Antigens, CD , Antigens, Differentiation/metabolism , Antigens, Heterophile/immunology , B7-1 Antigen/biosynthesis , B7-1 Antigen/genetics , Base Sequence , Blotting, Northern , CD28 Antigens/metabolism , CTLA-4 Antigen , Cells, Cultured , Cloning, Molecular , Dimerization , Humans , Immunosuppressive Agents/pharmacology , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , Isoantigens/immunology , Jurkat Cells , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Membrane Proteins/biosynthesis , Membrane Proteins/chemistry , Molecular Sequence Data , Protein Binding , Solubility , Swine , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
J Thorac Cardiovasc Surg ; 119(4 Pt 1): 690-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10733757

ABSTRACT

BACKGROUND: Pulmonary xenotransplantation is currently limited by hyperacute rejection mediated in part by xenoreactive natural antibody and complement. Transgenic swine organs that express the human complement regulatory protein CD59 have demonstrated improved survival in models of pig-to-primate xenotransplantation. OBJECTIVE: The purpose of this study was to evaluate transgenic swine lungs that express the human complement regulatory protein CD59 in a model of pig-to-human xenotransplantation. METHODS: Transgenic swine lungs (n = 5, experimental group) and outbred swine lungs (n = 6, control group) were perfused with fresh, whole human blood through a centrifugal pump on an ex vivo circuit. Functional data were collected throughout perfusion. Immunoglobulin and complement studies were performed on perfusate samples, and both histologic and immunofluorescent analyses were performed on tissue sections. RESULTS: Mean lung survival for the experimental group was increased when compared with controls, 240 +/- 0 minutes versus 35.3 +/- 14.5 minutes, respectively, with a P value of less than.01. A decreased rise in pulmonary vascular resistance at 15 minutes was observed in the experimental group (343 +/- 87 mm Hg. L(-1). min(-1), in contrast to the control group (1579 +/- 722 mm Hg. L(-1). min(-1); P <.01). Pulmonary compliance at 15 minutes was improved for the experimental group versus control group (9.31 +/- 1.41 mL. cm(-2) H(2)O and 4.11 +/- 2.84 mL. cm(-2) H(2)O, respectively; P <.01). SC5b-9 generation in the plasma perfusate was delayed for the experimental group versus the control group. Immunofluorescent examination of tissue sections demonstrated equivalent deposition of immunoglobulin G, immunoglobulin M, C1q, and C3 in both groups, with reduced deposition of C9 in the experimental group. CONCLUSIONS: Transgenic swine pulmonary xenografts that express the human complement regulatory protein CD59 demonstrated improved function and survival in an ex vivo model of pig-to-human xenotransplantation.


Subject(s)
CD59 Antigens/analysis , Graft Survival/immunology , Lung Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Complement C3a/analysis , Complement Hemolytic Activity Assay , Fluorescent Antibody Technique , Humans , Immunoglobulin G/analysis , Immunoglobulin M/analysis , In Vitro Techniques , Lung/immunology , Lung/pathology , Lung Compliance , Pulmonary Circulation , Swine , Vascular Resistance
7.
FASEB J ; 13(13): 1762-73, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10506579

ABSTRACT

Hyperacute rejection (HAR) is the first critical immunological hurdle that must be addressed in order to develop xenogeneic organs for human transplantation. In the area of cell-based xenotransplant therapies, natural antibodies (XNA) and complement have also been considered barriers to successful engraftment. Transgenic expression of human complement inhibitors in donor cells and organs has significantly prolonged the survival of xenografts. However, expression of complement inhibitors without eliminating xenogeneic natural antibody (XNA) reactivity may provide insufficient protection for clinical application. An approach designed to prevent XNA reactivity during HAR is the expression of human alpha1, 2-fucosyltransferase (H-transferase, HT). H-transferase expression modifies the cell surface carbohydrate phenotype of the xenogeneic cell, resulting in the expression of the universal donor O antigen and a concomitant reduction in the expression of the antigenic Galalpha1,3-Gal epitope. We have engineered various transgenic pig lines that express HT in different cells and tissues, including the vascular endothelium. We demonstrate that in two different HT transgenic lines containing two different HT promoter constructs, expression can be differentially regulated in a constitutive and cytokine-inducible manner. The transgenic expression of HT results in a significant reduction in the expression of the Galalpha1,3-Gal epitope, reduced XNA reactivity, and an increased resistance to human serum-mediated cytolysis. Transgenic pigs that express H-transferase promise to become key components for the development of xenogeneic cells and organs for human transplantation.


Subject(s)
Fucosyltransferases/biosynthesis , Graft Rejection/blood , Swine/genetics , Swine/immunology , Transplantation, Heterologous/immunology , ABO Blood-Group System/immunology , Animals , Animals, Genetically Modified , Aorta/immunology , Cell Membrane/immunology , Endothelium, Vascular/immunology , Fibroblasts/immunology , Fucosyltransferases/genetics , Humans , Membrane Glycoproteins/immunology , Phenotype , Galactoside 2-alpha-L-fucosyltransferase
8.
Xenotransplantation ; 6(1): 6-16, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10355728

ABSTRACT

Hyperacute rejection (HAR) remains a critical immunologic hurdle in the development of xenogeneic organs for human transplantation. Strategies that simultaneously eliminate both natural antibody reactivity and complement activation on the xenogeneic cell surface may be the best approach to achieve clinical application of xenogeneic vascularized organ transplantation. We have developed multiple lines of genetically manipulated mice to evaluate the combination of different genetic approaches aimed at inhibiting antibody and complement-mediated cell lysis. We utilized transgenic mice expressing the human complement inhibitor, CD59, the human 1,2-fucosyltransferase (H-transferase, HT) and the alpha1,3-galactosyltransferase (alpha1,3-GT) knock-out mouse line (Gal KO). Our data show that expression of hCD59 in combination with HT expression or the null phenotype of alpha1,3-GT are equally effective at preventing human serum-mediated cytolysis. Interestingly, the triple combination affords no additional protective effect. Therefore, coexpression of HT and a complement inhibitor is the most immediate strategy to genetically engineer transgenic pigs to be used as xenogeneic donors.


Subject(s)
Cytotoxicity, Immunologic/genetics , Graft Rejection/genetics , Animals , Antibodies, Heterophile/biosynthesis , Base Sequence , CD59 Antigens/genetics , Complement System Proteins/metabolism , DNA Primers/genetics , Fucosyltransferases/genetics , Galactosyltransferases/genetics , Humans , Immunity, Innate , Mice , Mice, Knockout , Mice, Transgenic , Transplantation, Heterologous , Galactoside 2-alpha-L-fucosyltransferase
9.
World J Surg ; 21(9): 951-5, 1997.
Article in English | MEDLINE | ID: mdl-9361510

ABSTRACT

The lack of sufficient suitable human donor lungs for the many patients requiring pulmonary transplantation as life-saving therapy for end-stage lung diseases has generated extensive interest in cross-species lung transplantation. Ethical concerns and those of animal rights advocates have prompted studies of nonprimate species as potential solid organ donors for humans. This paper provides an overview of some of the laboratory studies of cross-species pulmonary transplantation performed over the past 20 years and focuses, in particular, on more recent work (from our laboratory and others) in the area of porcine-to-primate pulmonary xenotransplantation.


Subject(s)
Lung Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Animals, Genetically Modified , CD59 Antigens/immunology , Disease Models, Animal , Graft Survival , Humans , Lung Transplantation/methods , Lung Transplantation/pathology , Papio , Respiratory Insufficiency/surgery , Swine , Transplantation, Heterologous/methods , Transplantation, Heterologous/pathology
10.
Transplantation ; 64(3): 495-500, 1997 Aug 15.
Article in English | MEDLINE | ID: mdl-9275118

ABSTRACT

BACKGROUND: In pig-to-primate transplantation, antibody-mediated hyperacute rejection is the consequence of binding of natural antibodies to Gal alpha(1,3)Gal on pig endothelium. The elimination of the Gal alpha(1,3)Gal antigen from pig cells should prevent hyperacute rejection. Using in vitro techniques, we have previously reported that using the alpha1,2-fucosyltransferase gene induces the preferential expression of H substance with a concomitant reduction in the expression of Gal alpha(1,3)Gal. The aim of the present study was to examine the effect of expressing the alpha1,2-fucosyltransferase gene in vivo on Gal alpha(1,3)Gal. METHODS: Three alpha1,2-fucosyltransferase transgenic lines of mice were produced and characterized serologically and histologically. RESULTS: Immunohistological studies showed heavy staining for H substance in liver, spleen, kidney, and heart, with a reduction in staining for Gal alpha(1,3)Gal. In addition, there was a reduction in the binding of human anti-Gal alpha(1,3)Gal antibody to lymphocytes from alpha1,2-fucosyltransferase transgenic mice and a substantial decrease in complement-mediated cytolysis of alpha1,2-fucosyltransferase transgenic lymphocytes when compared with that obtained with normal mice. CONCLUSIONS: The findings have important implications, in that alpha1,2-fucosyltransferase transgenic pigs could be produced as a source for humans. Such pigs should have a reduced expression of Gal alpha(1,3)Gal.


Subject(s)
Disaccharides/genetics , Fucosyltransferases/pharmacology , Mice, Transgenic/genetics , Plant Lectins , ABO Blood-Group System , Animals , Down-Regulation/drug effects , Female , Fucosyltransferases/chemistry , Gene Expression/drug effects , Gene Expression/physiology , Hemagglutinins/metabolism , Humans , Lectins/pharmacology , Mice , Mice, Inbred C57BL , Galactoside 2-alpha-L-fucosyltransferase
11.
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
14.
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
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.
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.
Proc Natl Acad Sci U S A ; 91(23): 11153-7, 1994 Nov 08.
Article in English | MEDLINE | ID: mdl-7526391

ABSTRACT

The serious shortage of human organs available for transplantation has engendered a heightened interest in the use of animal organs (xenografts) for transplantation. However, the major barrier to successful discordant xenogeneic organ transplantation is the phenomenon of hyperacute rejection. 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. Although endogenous membrane-associated complement inhibitors normally protect endothelial cells from autologous complement, they are species restricted and thus confer limited resistance to activated xenogeneic complement. To address the pathogenesis of hyperacute rejection in xenotransplantation, transgenic mice and a transgenic pig were engineered to express the human terminal complement inhibitor hCD59. High-level cell surface expression of hCD59 was achieved in a variety of murine and porcine cell types, most importantly on both large vessel and capillary endothelium. hCD59-expressing porcine cells were significantly resistant to challenge with high-titer anti-porcine antibody and human complement. These experiments demonstrate a strategy for developing a pig-to-primate xenogeneic transplantation model to test whether the expression of a human complement inhibitor in transgenic pigs could render xenogeneic organs resistant to hyperacute rejection.


Subject(s)
Antigens, CD/genetics , Complement Inactivator Proteins/genetics , Membrane Glycoproteins/genetics , Transplantation, Heterologous/methods , Animals , Animals, Genetically Modified , CD59 Antigens , Endothelium, Vascular/immunology , Genes , Graft Rejection/prevention & control , H-2 Antigens/genetics , Humans , Promoter Regions, Genetic , Swine
20.
J Virol ; 68(2): 730-7, 1994 Feb.
Article in English | MEDLINE | ID: mdl-7507185

ABSTRACT

Herpesvirus saimiri (HVS) is a lymphotropic herpesvirus that induces T-cell transformation in vitro and causes lymphomas and leukemias in New World primates other than its natural host, the squirrel monkey. Nucleotide sequence analysis of the HVS genome revealed two open reading frames with significant homology to genes for human complement regulatory molecules. One of these genes encodes a predicted protein (designated HVSCD59) with 48% amino acid sequence identity to the human terminal complement regulatory protein CD59 (HuCD59). The CD59 homolog from squirrel monkey (SMCD59) was cloned, and the corresponding amino acid sequence showed 69% identity with HVSCD59. BALB/3T3 cells stably expressing HVSCD59, SMCD59, or HuCD59 were equally protected from complement-mediated lysis by human serum. However, only HVSCD59-expressing cells were effectively protected from complement-mediated lysis when challenged with rat serum, suggesting that HVSCD59 was less species restrictive. The complement regulatory activity of HVSCD59 and SMCD59 occurred after C3b deposition, indicating terminal complement inhibition. Treatment of BALB/3T3 stable transfectants with phosphatidylinositol-specific phospholipase C prior to complement attack decreased the complement regulatory function of HVSCD59, suggesting cell surface attachment via a glycosyl-phosphatidylinositol anchor. Cells expressing HVSCD59 effectively inhibited complement-mediated lysis by squirrel monkey serum in comparison with SMCD59-expressing cells. Finally HVSCD59-specific transcripts were detected in owl monkey cells permissive for lytic HVS replication but not in T cells transformed by HVS, which failed to produce virions. These data are the first to demonstrate a functional, virally encoded terminal complement inhibitor and suggest that HVSCD59 represents a humoral immune evasion mechanism supporting the lytic life cycle of HVS.


Subject(s)
Antigens, CD/immunology , Complement Inactivator Proteins/immunology , Complement System Proteins/immunology , Herpesvirus 2, Saimiriine/immunology , Immunity, Cellular/immunology , Membrane Glycoproteins/immunology , Viral Proteins/immunology , 3T3 Cells , Amino Acid Sequence , Animals , Antigens, CD/genetics , Base Sequence , CD59 Antigens , Cell Death , Complement C3/metabolism , Complement Inactivator Proteins/biosynthesis , Complement Inactivator Proteins/drug effects , Complement Inactivator Proteins/genetics , Herpesvirus 2, Saimiriine/genetics , Humans , Membrane Glycoproteins/genetics , Mice , Molecular Sequence Data , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Phosphoric Diester Hydrolases/pharmacology , Rats , Saimiri/immunology , Sequence Homology, Amino Acid , Species Specificity , Transcription, Genetic , Viral Proteins/biosynthesis , Viral Proteins/drug effects , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...