Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Thorac Cardiovasc Surg ; 115(5): 998-1006, 1998 May.
Article in English | MEDLINE | ID: mdl-9605067

ABSTRACT

OBJECTIVE: Pig hearts transplanted into unmedicated newborn baboons do not undergo hyperacute rejection by preformed xenoantibody and complement. These grafts are rejected at days 3 to 4 in association with the infiltration of macrophages and natural killer cells. We investigated whether an immunosuppressive regimen used widely in cardiac allotransplantation could reduce this cellular response and prolong xenograft life. METHODS: Ten newborn baboons underwent heterotopic pig cardiac xenotransplantation. Five baboons were immunosuppressed with mycophenolate mofetil (100 mg/kg), methylprednisolone acetate (0.8 mg/kg), and cyclosporine A (INN: ciclosporin; 10 mg/kg). Xenograft rejection was studied by light microscopy and immunofluorescence. The induced humoral response to porcine xenoantigens was documented by enzyme-linked immunosorbent assay using synthetic alpha-1,3-galactosyl epitopes coupled to bovine serum albumin. RESULTS: Graft life was extended from a mean of 3.6 +/- 0.5 days (n = 5) to a mean of 6.2 +/- 1.1 days (n = 5, p = 0.01). In comparison with controls, explanted grafts from medicated baboons demonstrated reduced infiltration with natural killer cells and macrophages, but increased evidence of complement-mediated rejection substantiated by increased deposition of immunoglobulin M, complement, and fibrin. In all baboons receiving transplants, levels of both immunoglobulin M and immunoglobulin G anti-galactose were significantly increased after transplantation, with immunoglobulin G levels remaining persistently elevated. CONCLUSIONS: These results indicate that cyclosporine-based triple immunosuppression marginally prolonged xenograft survival and appears to have reduced the natural killer cell and macrophage infiltrates. The immunosuppressive protocol, however, was not adequate to prevent the induced immunoglobulin M humoral response and prevent complement-mediated graft injury.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Graft Survival/drug effects , Heart Transplantation , Immunosuppressive Agents/therapeutic use , Leukocytes, Mononuclear/immunology , Animals , Animals, Newborn , Complement C3/immunology , Cyclosporine/therapeutic use , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Female , Fibrin/metabolism , Heart Transplantation/immunology , Heart Transplantation/pathology , Immune Tolerance/drug effects , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Leukocytes, Mononuclear/pathology , Methylprednisolone/analogs & derivatives , Methylprednisolone/therapeutic use , Methylprednisolone Acetate , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/therapeutic use , Myocardium/pathology , Papio , Swine , Transplantation, Heterologous
2.
Int J Artif Organs ; 20(7): 375-8, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9298409

ABSTRACT

The purpose of this study was to evaluate effects of DL-Penicillamine (DLP), a compound interrupting S-S bonds (IgM pentamers) on binding and cytotoxicity of adult baboon performed xenoantibodies to pig endothelial cells. Pooled baboon serum was treated with different concentrations of DLP during various periods of time. Complement-mediated cytotoxicity assay was used to determine the reactivity of baboon xenoantibodies to pig aortic endothelial cells (PAEC). To assess IgM and IgG binding to PAEC, ELISA method was applied. Serum treated with DLP revealed significant reduction of cytotoxicity in a dose dependent manner. Cytotoxicity was also reduced during time prolongation of DLP exposure to PAEC. Results indicate that baboon performed IgM and IgG xenoantibodies bind to pig endothelial cells, but only IgM is able to cause degradation of the complement. DLP significantly reduces cytotoxicity and eliminates binding of IgMs to PAEC in spite of continued binding of IgG xenoantibodies to the surface of endothelium.


Subject(s)
Antibodies, Heterophile/drug effects , Immunoglobulin G/drug effects , Immunoglobulin M/drug effects , Penicillamine/pharmacology , Animals , Antibodies, Heterophile/metabolism , Binding, Competitive/drug effects , Cell Adhesion/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Papio , Stereoisomerism , Swine , Transplantation, Heterologous
3.
Curr Opin Cardiol ; 12(2): 172-8, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9192487

ABSTRACT

The severe shortage of human donor hearts has prompted several investigators to develop alternative strategies using cross-specie organs or xenografts. Unlike human allotransplantation, in which the important antigenic differences between donor and recipient are confined to the major histocompatibility and blood group antigens, xenotransplantation is confronted with the potential for multiple antigenic differences. In the pig-to-primate model of xenotransplantation, the primary obstacle to cross-species transplantation has been hyperacute rejection mediated by complement fixing antibodies directed against galactose alpha 1,3-galactose (Gal alpha 1,3-Gal) epitopes on the pig endothelium. Conventional immunosuppression is unable to overcome hyperacute rejection; however, recent efforts in molecular biology have focused on genetically engineering porcine donors to express human proteins in their tissue. Transgenic pigs that express human complement regulatory proteins on their endothelium have been developed. Heterotopic transplantation of these transgenic donor hearts have had only moderate success. Alternative approaches attempt to eliminate the Gal alpha 1,3-Gal epitopes by genetically "knocking out" the enzyme necessary for its synthesis, or to reduce the expression of Gal alpha 1,3-Gal epitopes by genetically inserting enzymes that redirect precursor molecules into alternative synthetic pathways. The technology to knock out the necessary enzymes in pigs is not yet available; however, pigs expressing the H-transferase gene have been developed and show reduced levels of Gal alpha 1,3-Gal epitopes. Although this "new breed" of transgenic pigs may overcome the barrier of hyperacute rejection, special strategies will need to be developed that target the next barrier of xenograft rejection.


Subject(s)
Graft Rejection/immunology , Heart Transplantation/methods , Transplantation, Heterologous , Animals , Antigen-Antibody Reactions , Graft Rejection/pathology , Graft Rejection/prevention & control , Heart Transplantation/immunology , Heart Transplantation/pathology , Histocompatibility/immunology , Humans , Immunosuppressive Agents/therapeutic use , Tissue Donors , Transplantation, Heterologous/immunology , Transplantation, Heterologous/pathology
4.
Ann Transplant ; 2(1): 14-20, 1997.
Article in English | MEDLINE | ID: mdl-9869836

ABSTRACT

We have previously described a form of xenograft rejection, mediated by natural killer (NK) cells, occurring in pig-to-primate organ transplants beyond the period of antibody-mediated hyperacute rejection. In this study, two distinct NK activation pathways were identified as mechanisms of pig aortic endotheliual cell (PAEC) lysis by human NK cells. Using an antibody-dependent cellular cytotoxicity (ADCC) assay, a progressive increase in human NK lysis of PAEC was observed following incubation with human IgG at increasing serum titer. In the absence of IgG, a second mechanism of PAEC lysis by human NK cells was observed following activation with IL-2. IL-2 activation of human NK cells increased lysis of PAEC by over 3-fold compared with ADCC. These results indicate that IL-2 activation of human NK cells induces significantly higher levels of lytic activity than does conventional ADCC involving IgG and FcRIII. We next investigated the role of MHC class I molecules in the regulation of NK lysis following IL-2 activation. PAEC expression of SLA class I molecules was increased by up to 75% by treatment with human TNFa. Following treatment with TNFa at 1 u/ml, IL-2 activated human NK lysis of PAEC was inhibited at every effector:target (E:T) ratio tested. Maximal effect occurred at an E:T ratio of 10:1, with TNFa inhibiting specific lysis by 59% (p < 0.01). Incubation with an anti-SLA class I Mab, but not IgG isotype control, abrogated the protective effects of TNFa on NK lysis of PAEC, suggesting direct inhibitory effects of SLA class I molecules on human NK function. To investigate whether human MHC class I molecules might have similar effects on human NK lysis of PAEC, further experiments were performed using a soluble peptide derived from the alpha-helical region of HLA-B7. Incubation with the HLA-B7 derived peptide significantly reduced the IL-2 activated NK lytic activity against PAEC in a dose-dependent fashion. Maximal effect occurred at a concentration of 10 mg/ml, where an 8-fold reduction in IL-2 augmented NK lysis was observed (p < 0.01). These results suggest that IL-2 activated human NK lysis of porcine xenografts may be inhibited by strategies which increase PAEC expression of SLA class I molecules, introduce HLA class I genes into PAEC, or use soluble HLA class I peptides.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Endothelium, Vascular/immunology , Histocompatibility Antigens Class I/immunology , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Aorta/immunology , Cells, Cultured , Graft Rejection/immunology , HLA-B7 Antigen/immunology , Humans , Killer Cells, Natural/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/drug effects , Swine , Transplantation, Heterologous/immunology , Tumor Necrosis Factor-alpha/pharmacology
5.
J Thorac Cardiovasc Surg ; 112(4): 1002-9, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8873727

ABSTRACT

The humoral and cell-mediated immune responses to subsequent allografts were determined in primate recipients after concordant xenotransplantation as a bridge to allotransplantation. Heterotopic heart transplants (n = 4) were performed from cynomolgus monkeys into ABH type-matched olive baboons followed 2 weeks later by allotransplantation from ABH type-matched baboon donors. Allografts were explanted at 8 weeks. All recipients underwent splenectomy at the time of xenotransplantation and received immunosuppression with cyclosporine, azathioprine, and methylprednisolone. Concordant xenotransplantation in these primates did not induce humoral or cell-mediated immune responses that jeopardized subsequent allografts. The degree of xenospecific immune reactivity, as determined by specific cytotoxicity of recipient T-cell lines derived from the xenograft and extent of histologic xenograft rejection, did not predict the severity of subsequent allograft rejection. In two of the four recipients, xenotransplantation induced an alloreactive humoral response against antigens expressed by the B cells of more than 50% of members from a panel of 12 unrelated baboons. In all recipients, priming with xenogeneic splenocytes in vitro induced an accelerated proliferative T-cell response to allogeneic lymphocytes from 16% of this panel. This study affirms the role of concordant xenografts as appropriate biologic bridges to human allotransplantation. However, our results suggest that xenoreactive baboon memory CD4 T cells may recognize major histocompatibility complex class II--like structures shared between the xenogeneic and allogeneic targets. The potential allorecognition induced by a xenograft may affect the process of subsequent allograft donor selection.


Subject(s)
Transplantation, Heterologous/immunology , Transplantation, Homologous/immunology , Animals , Antibody Formation , Cytotoxicity, Immunologic , Graft Rejection , Heart Transplantation/immunology , Histocompatibility Antigens Class II/immunology , In Vitro Techniques , Lymphocyte Activation , Macaca fascicularis , Papio , T-Lymphocytes/immunology
9.
Transplantation ; 61(3): 356-63, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8610340

ABSTRACT

The interaction of T cell costimulatory molecules with their ligands is required for optimal T cell activation. Interference with such interactions can induce antigen unresponsiveness and delay xeno- and allograft rejection. We have previously shown that LFA3TIP, a soluble human lymphocyte function-associated antigen (LFA)-3 construct, binds CD2 and inhibits responses of human T cells in vitro. This study reports the first use of a human fusion protein, LFA3TIP, to significantly prolong primate cardiac allograft survival. Based on our observations that LFA3TIP inhibits baboon allogeneic mixed lymphocyte reactions, we gave baboon recipients of heterotopic cardiac allografts injections of LFA3TIP, 3 mg/kg i.v., for 12 consecutive days, starting 2 days before transplantation. This regimen delayed graft rejection from an average of 10.6 +/- 2.3 days for human IgG-treated controls (n = 5) to an average of 18.0 +/- 5.3 days for LFA3TIP-injected animals (n = 7; P < or = 0.01). Grafts from LFA3TIP-treated animals showed markedly diminished coronary endothelialitis as compared with control animals. LFA3TIP reached peak serum levels of approximately 100 micrograms/ml after 7-9 injections and persisted in the 10-micrograms/ml range for 1 to 2 weeks after the final injection. Despite these blood levels, circulating antibodies to LFA3TIP were not detected in the serum. No renal or hepatic toxicity was noted. The possible mechanism by which LFA3TIP acts to inhibit graft rejection is discussed; success in prolonging graft survival when LFA3TIP is used as a single-agent therapy suggests great potential for this novel therapeutic agent.


Subject(s)
CD58 Antigens/therapeutic use , Heart Transplantation/immunology , Immunoglobulin G/therapeutic use , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/therapeutic use , CD2 Antigens/metabolism , CD58 Antigens/administration & dosage , CD58 Antigens/blood , Graft Rejection/prevention & control , Graft Survival , Heart Transplantation/adverse effects , Heart Transplantation/pathology , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/blood , Immunotherapy , In Vitro Techniques , Lymphocyte Culture Test, Mixed , Papio , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/therapeutic use , Time Factors , Transplantation, Heterotopic , Transplantation, Homologous
11.
J Thorac Cardiovasc Surg ; 111(1): 13-8, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8551757

ABSTRACT

We investigated the efficacy of the Jarvik 2000 intraventricular assist device (Jarvik Research, Inc., New York, N.Y.) in an ovine model. The device is an axial flow pump measuring 1.8 cm in diameter by 5 cm long, has a displacement volume of 12 ml, and can deliver flow from 2 to 7 L/min. Seven devices were implanted through a left thoracotomy into the left ventricle with an outflow graft to the descending aorta. Animals were treated with warfarin sodium and aspirin to maintain prothrombin times approximately 1.5 times control. Animals were followed up for 3 to 123 days. Two animals died of operative complications at days 3 and 5. One device failed at 58 days because of thrombus formation at the inflow side of the impeller. The remaining four animals were killed at days 19, 42, 42, and 123, respectively, because of broken electric power cables. Hematocrit values rose significantly higher than preoperative levels (22.8% +/- 3.8% to 30.5% +/- 3.4%); premortem elevations of values higher than baseline values of plasma free hemoglobin (10.4 +/- 7.8 mg/dl to 17.1 +/- 7.4 mg/dl) and lactate dehydrogenase (391.5 +/- 113.7 units/L to 771.2 +/- 370.8 units/L) were statistically insignificant. Serum creatinine and bilirubin levels were normal. No end-organ dysfunction arising from long-term support was evident clinically or at postmortem examination, nor was there any evidence of embolism or damage to intracardiac structures. We found the Jarvik 2000 intraventricular assist device to be easily implantable, safe, nonhemolytic, and able to provide physiologic flow with power requirements under 10 watts.


Subject(s)
Heart-Assist Devices , Adult , Animals , Anticoagulants/therapeutic use , Aspirin/therapeutic use , Blood Vessel Prosthesis , Child , Coronary Circulation/physiology , Electric Power Supplies , Embolism/prevention & control , Equipment Design , Heparin/therapeutic use , Humans , Polyethylene Terephthalates , Sheep , Thrombosis/prevention & control , Warfarin/therapeutic use
12.
Transplantation ; 59(3): 410-6, 1995 Feb 15.
Article in English | MEDLINE | ID: mdl-7871572

ABSTRACT

The shortage of organ donors for transplantation is more pronounced for the lung than for any other solid organ. To address this problem, we evaluated the feasibility of pulmonary xenotransplantation. Preliminary investigations demonstrated that orthotopically placed pig lungs in cynomologous monkey recipients could be engrafted up to 9 hr after reperfusion without evidence of hyperacute rejection. In this study, the rejection reaction of pig lungs transplanted orthotopically into baboons (n = 6) was further investigated by ELISA and immunohistochemistry. Four baboon recipients were killed at 24 hr and 2 recipients were killed at 72 hr after transplantation. Pulmonary arterial flow measurements demonstrated flow to the grafts, and systemic arterial and xenograft pulmonary venous blood gas analysis suggested function of the donor lungs during the course of engraftment. Serum levels of baboon anti-pig endothelial cell xenoantibody were normal and decreased minimally over time. Immunohistochemical staining of biopsies demonstrated trace IgG and IgM along graft endothelium 2 hr after reperfusion. At 8 hr, biopsy samples showed no immunoglobulin bound to endothelial cells. Staining for complement was negative. Fibrin and platelets were detected along xenograft endothelium. Despite these findings, the lung xenografts appeared injured and clinically rejected. During the first 8 hr after reperfusion, the grafts were hyperemic and subsequently became focally ecchymotic. Chest x-rays showed progressive pulmonary congestion. These findings suggest that the lung may be relatively resistant to antibody-mediated hyperacute rejection and efforts are being directed toward identifying the mechanism of the observed xenograft lung injury.


Subject(s)
Graft Rejection/physiopathology , Lung Transplantation , Animals , Animals, Newborn , Antibodies, Heterophile/blood , Biopsy , Blood Gas Analysis , Endothelium/immunology , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Immunohistochemistry , Lung/diagnostic imaging , Lung/physiopathology , Papio , Radiography , Regional Blood Flow/physiology , Swine , Transplantation, Heterologous , X-Rays
13.
Transplantation ; 59(1): 1-6, 1995 Jan 15.
Article in English | MEDLINE | ID: mdl-7839407

ABSTRACT

The shortage of organs for transplantation is especially severe for the critically ill newborn infant, for whom donors of the appropriate size are particularly scarce. One way to overcome this problem is to use animals in lieu of humans as organ donors. The major limitation to using animals for this purpose is the susceptibility of animal organs to hyperacute rejection, a violent rejection reaction thought to be mediated by antidonor antibody and complement. To evaluate the potential application of xenotransplantation to newborns, we tested neonatal humans and neonatal baboons and found that neither population expressed significant levels of xenoreactive anti-pig antibodies. We transplanted heterotopically hearts from newborn pigs into unmanipulated newborn baboons (n = 4). There was no evidence of hyperacute rejection in any of the grafts; the animals were killed with functioning grafts at 15, 81, 82, and 82 hr. This outcome contrasts with that of newborn pig-to-mature baboon and mature pig-to-mature baboon cardiac xenografts, which were rejected within 1 hr of transplantation. The histology of pig graft biopsies from the newborn recipients was normal. Immunohistochemistry revealed only traces of IgM, C3, C4, and the membrane attack complex along graft endothelium. Fibrin deposition along endothelial surfaces was observed early after transplantation and became more extensive with time; in the absence of endothelial bound antibody or complement, this change may represent preservation injury. This study suggests that due to low levels of natural antibody, the newborn infant may permit prolonged xenograft survival.


Subject(s)
Animals, Newborn/immunology , Graft Rejection/immunology , Heart Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Complement C3/analysis , Complement C4/analysis , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Fibrin/analysis , Graft Rejection/pathology , Graft Survival/immunology , Heart Transplantation/pathology , Humans , Immunoglobulin M/analysis , Immunoglobulin M/blood , Immunohistochemistry , Infant, Newborn , Papio , Swine , Time Factors , Transplantation, Heterologous/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...