Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Xenotransplantation ; 31(3): e12865, 2024.
Article in English | MEDLINE | ID: mdl-38853364

ABSTRACT

Recent clinical xenotransplantation and human decedent studies demonstrate that clinical hyperacute rejection of genetically engineered porcine organs can be reliably avoided but that antibody mediated rejection (AMR) continues to limit graft survival. We previously identified porcine glycans and proteins which are immunogenic after cardiac xenotransplantation in non-human primates, but the clinical immune response to antigens present in glycan depleted triple knockout (TKO) donor pigs is poorly understood. In this study we use fluorescence barcoded human embryonic kidney cells (HEK) and HEK cell lines expressing porcine glycans (Gal and SDa) or proteins (tetraspanin-29 [CD9], membrane cofactor protein [CD46], protectin, membrane attack complex inhibition factor [CD59], endothelial cell protein C receptor, and Annexin A2) to screen antibody reactivity in human serum from 160 swine veterinarians, a serum source with potential occupational immune challenge from porcine tissues and pathogens. High levels of anti-Gal IgM were present in all samples and lower levels of anti-SDa IgM were present in 41% of samples. IgM binding to porcine proteins, primarily CD9 and CD46, previously identified as immunogenic in pig to non-human primate cardiac xenograft recipients, was detected in 28 of the 160 swine veterinarian samples. These results suggest that barcoded HEK cell lines expressing porcine protein antigens can be useful for screening human patient serum. A comprehensive analysis of sera from clinical xenotransplant recipients to define a panel of commonly immunogenic porcine antigens will likely be necessary to establish an array of porcine non-Gal antigens for effective monitoring of patient immune responses and allow earlier therapies to reverse AMR.


Subject(s)
Graft Rejection , Transplantation, Heterologous , Animals , Transplantation, Heterologous/methods , Humans , Swine , Graft Rejection/immunology , HEK293 Cells , Veterinarians , Polysaccharides/immunology , Animals, Genetically Modified , Antibodies, Heterophile/immunology , Antibodies, Heterophile/blood , Heterografts/immunology , Immunoglobulin M/immunology , Immunoglobulin M/blood
2.
J Thorac Cardiovasc Surg ; 166(4): e142-e152, 2023 10.
Article in English | MEDLINE | ID: mdl-36914518

ABSTRACT

BACKGROUND: Heart valve implantation in juvenile sheep to demonstrate biocompatibility and physiologic performance is the accepted model for regulatory approval of new biological heart valves (BHVs). However, this standard model does not detect the immunologic incompatibility between the major xenogeneic antigen, galactose-α-1,3-galactose (Gal), which is present in all current commercial BHVs, and patients who universally produce anti-Gal antibody. This clinical discordance leads to induced anti-Gal antibody in BHV recipients, promoting tissue calcification and premature structural valve degeneration, especially in young patients. The objective of the present study was to develop genetically engineered sheep that, like humans, produce anti-Gal antibody and mirror current clinical immune discordance. METHODS: Guide RNA for CRISPR Cas9 nuclease was transfected into sheep fetal fibroblasts, creating a biallelic frame shift mutation in exon 4 of the ovine α-galactosyltransferase gene (GGTA1). Somatic cell nuclear transfer was performed, and cloned embryos were transferred to synchronized recipients. Cloned offspring were analyzed for expression of Gal antigen and spontaneous production of anti-Gal antibody. RESULTS: Two of 4 surviving sheep survived long-term. One of the 2 was devoid of the Gal antigen (GalKO) and expressed cytotoxic anti-Gal antibody by age 2 to 3 months, which increased to clinically relevant levels by 6 months. CONCLUSIONS: GalKO sheep represent a new, clinically relevant advanced standard for preclinical testing of BHVs (surgical or transcatheter) by accounting for the first time for human immune responses to residual Gal antigen that persists after current BHV tissue processing. This will identify the consequences of immune disparity preclinically and avoid unexpected past clinical sequelae.


Subject(s)
Bioprosthesis , Calcinosis , Heart Valve Prosthesis , Animals , Humans , Sheep , Infant , Galactose , Heart Valves , Genetic Engineering
3.
Xenotransplantation ; 25(3): e12411, 2018 05.
Article in English | MEDLINE | ID: mdl-29913037

ABSTRACT

The major histocompatibility complex class I and class II human leukocyte antigens (HLA) play a central role in adaptive immunity but are also the dominant polymorphic proteins targeted in allograft rejection. Sensitized patients with high levels of panel-reactive anti-HLA antibody (PRA) are at risk of early allograft injury, rejection, reduced allograft survival and often experience prolonged waiting times prior to transplantation. Xenotransplantation, using genetically modified porcine organs, offers a unique source of donor organs for these highly sensitized patients if the anti-HLA antibody, which places the allograft at risk, does not also enhance anti-pig antibody reactivity responsible for xenograft rejection. Recent improvements in xenotransplantation efficacy have occurred due to improved immune suppression, identification of additional xenogeneic glycans, and continued improvements in donor pig genetic modification. Genetically engineered pig cells, devoid of the known xenogeneic glycans, minimize human antibody reactivity in 90% of human serum samples. For waitlisted patients, early comparisons of patient PRA and anti-pig antibody reactivity found no correlation suggesting that patients with high PRA levels were not at increased risk of xenograft rejection. Subsequent studies have found that some, but not all, highly sensitized patients express anti-HLA class I antibody which cross-reacts with swine leukocyte antigen (SLA) class I proteins. Recent detailed antigen-specific analysis suggests that porcine-specific anti-SLA antibody from sensitized patients binds cross-reactive groups present in a limited subset of HLA antigens. This suggests that using modern genetic methods, a program to eliminate specific SLA alleles through donor genetic engineering or stringent donor selection is possible to minimize recipient antibody reactivity even for highly sensitized individuals.


Subject(s)
Cross Reactions/immunology , HLA Antigens/immunology , Histocompatibility Antigens Class I/immunology , Leukocytes/immunology , Transplantation, Heterologous , Animals , Humans , Isoantibodies/immunology
4.
5.
J Immunol Res ; 2017: 2534653, 2017.
Article in English | MEDLINE | ID: mdl-29238731

ABSTRACT

Cardiac xenotransplantation (CXTx) is a promising solution to the chronic shortage of donor hearts. Recent advancements in immune suppression have greatly improved the survival of heterotopic CXTx, now extended beyond 2 years, and life-supporting kidney XTx. Advances in donor genetic modification (B4GALNT2 and CMAH mutations) with proven Gal-deficient donors expressing human complement regulatory protein(s) have also accelerated, reducing donor pig organ antigenicity. These advances can now be combined and tested in life-supporting orthotopic preclinical studies in nonhuman primates and immunologically appropriate models confirming their efficacy and safety for a clinical CXTx program. Preclinical studies should also allow for organ rejection to develop xenospecific assays and therapies to reverse rejection. The complexity of future clinical CXTx presents a substantial and unique set of regulatory challenges which must be addressed to avoid delay; however, dependent on these prospective life-supporting preclinical studies in NHPs, it appears that the scientific path forward is well defined and the era of clinical CXTx is approaching.


Subject(s)
Communicable Diseases/etiology , Heart Transplantation , Postoperative Complications/prevention & control , Animals , Communicable Disease Control , Genetic Therapy , Government Regulation , Heart Transplantation/legislation & jurisprudence , Heart Transplantation/methods , Humans , Immunosuppression Therapy , Mixed Function Oxygenases/genetics , N-Acetylgalactosaminyltransferases/genetics , Primates , Swine , Tissue Donors , Transplantation, Heterologous/legislation & jurisprudence , Transplantation, Heterologous/methods
6.
Xenotransplantation ; 22(6): 458-67, 2015.
Article in English | MEDLINE | ID: mdl-26490445

ABSTRACT

BACKGROUND: Rejection of Gal-free (GTKO) donor pig cardiac xenografts is strongly associated with vascular non-Gal antibody binding, endothelial cell (EC) injury, and activation and microvascular thrombosis. We adopted a pig-to-SCID/beige small animal transplant model to compare the pathogenicity of baboon and human anti-pig antibody. METHODS: Wild-type (GT(+) ) or GTKO porcine coronary arteries (PCAs) were transplanted into the infrarenal aorta of SCID/beige mice. Three days after transplant, recipients were infused with anti-pig antibody (anti-SLA class I, an isotype control, naive or sensitized baboon serum, or naive human serum). PCAs were recovered 24 h after antibody infusion and examined using histology, immunohistochemistry, and in situ hybridization. RESULTS: Dose-dependent intragraft thrombosis occurred after infusion of anti-SLA I antibody (but not isotype control) in GT(+) and GTKO PCA recipients. Naive baboon serum induced thrombosis in GT(+) grafts. Thrombosis was significantly reduced by pre-treating naive baboon serum with Gal polymer and not observed when this serum was infused to GTKO PCA recipients. Naive human serum caused dose-dependent intragraft thrombosis of GTKO PCAs. In all cases, thrombosis involved graft-specific vascular antibody and complement deposition, macrophage adherence, EC delamination, and subendothelial thrombus formation. CONCLUSIONS: This study provides the first direct in vivo comparison of the pathogenicity of naive human and baboon serum. The results suggest that human preformed non-Gal antibody may have increased pathogenicity compared to baboon. This model, which showed a rejected graft histopathology similar to antibody-mediated rejection in cardiac xenotransplantation, may be useful to assess the pathogenicity of individual protein or carbohydrate specific non-Gal reactive antibodies.


Subject(s)
Antibodies/immunology , Coronary Vessels/transplantation , Graft Rejection/immunology , Heterografts/transplantation , Papio/immunology , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Graft Survival/immunology , Humans , Mice, SCID , Swine , Transplantation, Heterologous/methods
7.
Int J Surg ; 23(Pt B): 223-228, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26306769

ABSTRACT

Genetic engineering of donor pigs to eliminate expression of the dominant xenogeneic antigen galactose α1,3 galactose (Gal) has created a sea change in the immunobiology of xenograft rejection. Antibody mediated xenograft rejection of GGTA-1 α-galactosyltransferase (GTKO) deficient organs is now directed to a combination of non-Gal pig protein and carbohydrate antigens. Glycan analysis of GTKO tissues identified no new neo-antigens but detected high levels of N-acetylneuraminic acid (Neu5Gc) modified glycoproteins and glycolipids. Humans produce anti-Neu5Gc antibody and in very limited clinical studies sometimes show an induced anti-Neu5Gc antibody response after challenge with pig tissue. The pathogenicity of anti-Neu5Gc antibody in xenotransplantation is not clear however as non-human transplant models, critical for modelling anti-Gal immunity, do not produce anti-Neu5Gc antibody. Antibody induced after xenotransplantation in non-human primates is directed to an array of pig endothelial cells proteins and to a glycan produced by the pig B4GALNT2 gene. We anticipate that immune suppression will significantly affect the T-cell dependent and independent specificity of an induced antibody response and that donor pigs deficient in synthesis of multiple xenogeneic glycans will be important to future studies.


Subject(s)
Antibodies/metabolism , Graft Rejection/immunology , Swine/immunology , Transplantation, Heterologous , Transplants/immunology , Animals , Animals, Genetically Modified/immunology , Disaccharides/immunology , Disaccharides/metabolism , Genetic Engineering/methods , Humans , N-Acetylneuraminic Acid/immunology , N-Acetylneuraminic Acid/metabolism , Polysaccharides/immunology , Primates/immunology , Swine/genetics
8.
Int J Surg ; 23(Pt B): 234-239, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26318967

ABSTRACT

Significant progress in understanding and overcoming cardiac xenograft rejection using a clinically relevant large animal pig-to-baboon model has accelerated in recent years. This advancement is based on improved immune suppression, which attained more effective regulation of B lymphocytes and possibly newer donor genetics. These improvements have enhanced heterotopic cardiac xenograft survival from a few weeks to over 2 years, achieved intrathoracic heterotopic cardiac xenograft survival of 50 days and orthotopic survival of 57 days. This encouraging progress has rekindled interest in xenotransplantation research and refocused efforts on preclinical orthotopic cardiac xenotransplantation.


Subject(s)
Graft Survival/immunology , Heart Transplantation/methods , Papio/immunology , Swine/genetics , Transplantation, Heterologous/methods , Animals , Immunosuppression Therapy/methods , Swine/immunology
9.
Xenotransplantation ; 21(6): 543-54, 2014.
Article in English | MEDLINE | ID: mdl-25176027

ABSTRACT

BACKGROUND: Xenograft rejection of pigs organs with an engineered mutation in the GGTA-1 gene (GTKO) remains a predominantly antibody mediated process which is directed to a variety of non-Gal protein and carbohydrate antigens. We previously used an expression library screening strategy to identify six porcine endothelial cell cDNAs which encode pig antigens that bind to IgG induced after pig-to-primate cardiac xenotransplantation. One of these gene products was a glycosyltransferase with homology to the bovine ß1,4 N-acetylgalactosaminyltransferase (B4GALNT2). We now characterize the porcine B4GALNT2 gene sequence, genomic organization, expression, and functional significance. METHODS: The porcine B4GALNT2 cDNA was recovered from the original library isolate, subcloned, sequenced, and used to identify a bacterial artificial chromosome (BAC) containing the entire B4GALNT2 locus from the Children's Hospital Oakland Research Institute BACPAC Resource Centre (#AC173453). PCR primers were designed to map the intron/exon genomic organization in the BAC clone. A stable human embryonic kidney (HEK) cell line expressing porcine B4GALNT2 (HEK-B4T) was produced. Expression of porcine B4GALNT2 in HEK-B4T cells was characterized by immune staining and siRNA transfection. The effects of B4GALNT2 expression in HEK-B4T cells was measured by flow cytometry and complement mediated lysis. Antibody binding to HEK and HEK-B4T cells was used to detect an induced antibody response to the B4GALNT2 produced glycan and the results were compared to GTKO PAEC specific non-Gal antibody induction. Expression of porcine B4GALNT2 in pig cells and tissues was measured by qualitative and quantitative real time reverse transcriptase PCR and by Dolichos biflorus agglutinin (DBA) tissue staining. RESULTS: The porcine B4GALNT2 gene shares a conserved genomic organization and encodes an open reading frame with 76 and 70% amino acid identity to the human and murine B4GALNT2 genes, respectively. The B4GALNT2 gene is expressed in porcine endothelial cells and shows a broadly distributed expression pattern. Expression of porcine B4GALNT2 in human HEK cells (HEK-B4T) results in increased binding of antibody to the B4GALNT2 enzyme, and increased reactivity with anti-Sd(a) and DBA. HEK-B4T cells show increased sensitivity to complement mediated lysis when challenged with serum from primates after pig to primate cardiac xenotransplantation. In GTKO and GTKO:CD55 cardiac xenotransplantation recipients there is a significant correlation between the induction of a non-Gal antibody, measured using GTKO PAECs, and the induction of antibodies which preferentially bind to HEK-B4T cells. CONCLUSION: The functional isolation of the porcine B4GALNT2 gene from a PAEC expression library, the pattern of B4GALNT2 gene expression and its sensitization of HEK-B4T cells to antibody binding and complement mediated lysis indicates that the enzymatic activity of porcine B4GALNT2 produces a new immunogenic non-Gal glycan which contributes in part to the non-Gal immune response detected after pig-to-baboon cardiac xenotransplantation.


Subject(s)
Antibodies/immunology , Graft Rejection/immunology , N-Acetylgalactosaminyltransferases/genetics , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Cell Line , Cells, Cultured , Cloning, Organism/methods , Humans , Papio/immunology , Swine , Transplantation, Heterologous/methods
10.
Xenotransplantation ; 21(6): 555-66, 2014.
Article in English | MEDLINE | ID: mdl-25176173

ABSTRACT

BACKGROUND: Scientists working in the field of xenotransplantation do not employ a uniform method to measure and report natural and induced antibody responses to non-Galα(1,3)Gal (non-Gal) epitopes. Such humoral responses are thought to be particularly pathogenic after transplantation of vascularized GalTKO pig organs and having a more uniform assay and reporting format would greatly facilitate comparisons between laboratories. METHODS: Flow cytometry allows examination of antibody reactivity to intact antigens in their natural location and conformation on cell membranes. We have established a simple and reproducible flow cytometric assay to detect antibodies specific for non-Gal pig antigens using primary porcine aortic endothelial cells (pAECs) and cell culture-adapted pAEC cell lines generated from wild type and α1,3galactosyl transferase knockout (GalTKO) swine. RESULTS: The consensus protocol we propose here is based on procedures routinely used in four xenotransplantation centers and was independently evaluated at three sites using shared cells and serum samples. Our observation support use of the cell culture-adapted GalTKO pAEC KO:15502 cells as a routine method to determine the reactivity of anti-non-Gal antibodies in human and baboon serum. CONCLUSIONS: We have developed an assay that allows the detection of natural and induced non-Gal xenoreactive antibodies present in human or baboon serum in a reliable and consistent manner. This consensus assay and format for reporting the data should be accessible to laboratories and will be useful for assessing experimental results between multiple research centers. Adopting this assay and format for reporting the data should facilitate the detection, monitoring, and detailed characterization of non-Gal antibody responses.


Subject(s)
Antibodies/pharmacology , Aorta/immunology , Endothelial Cells/immunology , Graft Rejection/immunology , Transplantation, Heterologous , Animals , Antibodies/immunology , Consensus , Endothelial Cells/metabolism , Graft Rejection/therapy , Immunoglobulins/immunology , Papio/immunology , Swine , Transplantation, Heterologous/methods
12.
J Heart Valve Dis ; 22(3): 383-90, 2013 May.
Article in English | MEDLINE | ID: mdl-24151765

ABSTRACT

BACKGROUND AND AIM OF THE STUDY: Current biological heart valves (BHVs) contain the major xenogeneic antigen Gal. Recipient anti-Gal antibody binding to such an implanted BHV may contribute to valve degeneration. The study aim was to compare, by implantation in non-human primates, the immune consequences of BHVs from Gal-positive wild-type (WT) pigs and those from alpha-galactosyltransferase knockout (GTKO) pigs. METHODS: Recipients were immunized prior to implant with keyhole limpet hemocyanin (KLH) conjugated to alphaGal to match the anti-Gal levels and isotypes found in humans. Stented glutaraldehyde-fixed BHVs from WT (n = 4) and GTKO (n = 3) pigs were commercially manufactured and implanted in the mitral position in non-human primates. Recipients were treated with enoxaparin (1 mg/kg b.i.d.) for five weeks which was tapered, and then discontinued. Serum antibody levels to Gal and KLH were measured using ELISA. RESULTS: Overall anti-Gal and anti-KLH antibody levels were decreased in both WT and GTKO BHV recipients after implantation. Serum anti-Gal IgG levels in GTKO BHV recipients fell rapidly within one month, matching the loss of anti-KLH reactivity. There was no significant difference in retention of anti-KLH antibody between the groups. WT BHV recipients retained significantly elevated levels of anti-Gal IgG during the first year post implant. Area under the curve analysis showed that anti-Gal IgG was significantly increased in the WT BHV group compared to GTKO BHV recipients (p < 0.01). CONCLUSION: Persistent and significantly (p < 0.01) elevated levels of anti-Gal IgG were observed in WT but not GTKO BHV non-human primate recipients, and indicated a continuing BHV-specific immune stimulation to the alphaGal antigen. These data support the hypothesis that the clinical use of Gal-positive xenogeneic bioprosthetic materials can induce an anti-Gal antibody response. Bioprosthetic devices prepared from GTKO pig tissue would eliminate immune stimulation to this major xenoreactive antigen, which may reduce the potential of immune-mediated injury and degeneration.


Subject(s)
Bioprosthesis , Disaccharides/immunology , Heart Valve Prosthesis Implantation , Heart Valve Prosthesis , Mitral Valve , Postoperative Complications , Trisaccharides/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Heterophile/immunology , Gene Knockout Techniques , Heart Valve Prosthesis Implantation/adverse effects , Heart Valve Prosthesis Implantation/methods , Hemocyanins/administration & dosage , Mitral Valve/immunology , Mitral Valve/transplantation , Models, Animal , Models, Cardiovascular , Models, Immunological , Monitoring, Immunologic , Papio , Postoperative Complications/immunology , Postoperative Complications/prevention & control
13.
Xenotransplantation ; 20(5): 292-307, 2013.
Article in English | MEDLINE | ID: mdl-25098626

ABSTRACT

The histopathology of cardiac xenograft rejection has evolved over the last 20 yr with the development of new modalities for limiting antibody-mediated injury, advancing regimens for immune suppression, and an ever-widening variety of new donor genetics. These new technologies have helped us progress from what was once an overwhelming anti-Gal-mediated hyperacute rejection to a more protracted anti-Gal-mediated vascular rejection to what is now a more complex manifestation of non-Gal humoral rejection and coagulation dysregulation. This review summarizes the changing histopathology of Gal- and non-Gal-mediated cardiac xenograft rejection and discusses the contributions of immune-mediated injury, species-specific immune-independent factors, transplant and therapeutic procedures, and donor genetics to the overall mechanism(s) of cardiac xenograft rejection.


Subject(s)
Antibodies/immunology , Graft Rejection/immunology , Animals , Graft Survival/immunology , Heart Transplantation/methods , Heterografts/immunology , Humans , Immunosuppression Therapy , Swine , Transplantation, Heterologous/methods
14.
Transplantation ; 93(7): 686-92, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22391577

ABSTRACT

BACKGROUND: Transgenic expression of human complement regulatory proteins reduces the frequency of hyperacute rejection (HAR) in Gal-positive cardiac xenotransplantation. In this study, we examined the impact of human CD55 (hCD55) expression on a Gal knockout (GTKO) background using pig-to-primate heterotopic cardiac xenotransplantation. METHODS: Cardiac xenotransplantation was performed with GTKO (group 1; n=6) and GTKO.hCD55 (group 2; n=5) donor pigs using similar immunosuppression. Cardiac biopsies were obtained 30 min after organ reperfusion. Rejection was characterized by histology and immunohistology. Intragraft gene expression, serum non-Gal antibody, and antibody recovered from rejected hearts were analyzed. RESULTS: HAR of a GTKO heart was observed. Remaining grafts developed delayed xenograft rejection. Median survival was 21 and 28 days for groups 1 and 2, respectively. Vascular antibody deposition was uniformly detected 30 min after organ reperfusion and at explant. A higher frequency of vascular C5b deposition was seen in GTKO organs at explant. Serum non-Gal antibody, antibody recovered from the graft, and intragraft gene expression were similar between the groups. CONCLUSION: HAR of GTKO hearts without hCD55 may occur. Expression of hCD55 seemed to restrict local complement activation but did not improve graft survival. Chronic vascular antibody deposition with evidence of protracted endothelial cell activation was seen. These observations suggest that non-Gal antibody-induced chronic endothelial cell activation coupled to possible hemostatic incompatibilities may be the primary stimulus for delayed xenograft rejection of GTKO hearts. To avoid possible HAR, future clinical studies should use donors expressing human complement regulatory proteins in the GTKO background.


Subject(s)
CD55 Antigens/metabolism , Complement Activation , Graft Rejection/prevention & control , Graft Survival , Heart Transplantation/immunology , Myocardium/immunology , Acute Disease , Animals , Animals, Genetically Modified , Biopsy , CD55 Antigens/genetics , Complement Activation/drug effects , Disaccharides/deficiency , Disaccharides/immunology , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Gene Knockout Techniques , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/drug effects , Humans , Immunohistochemistry , Immunosuppressive Agents/pharmacology , Myocardium/pathology , Papio , Swine , Time Factors , Transplantation, Heterologous
15.
Curr Opin Organ Transplant ; 17(2): 148-54, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22327911

ABSTRACT

PURPOSE OF REVIEW: Cardiac xenotransplantation (CXTx) remains a promising approach to alleviate the chronic shortage of donor hearts. This review summarizes recent results of heterotopic and orthotopic CXTx, highlights the role of non-Gal antibody in xenograft rejection, and discusses challenges to clinical orthotopic CXTx. RECENT FINDINGS: Pigs mutated in the α 1,3 galactosyltransferase gene (GTKO pigs) are devoid of the galactose α1,3 galactose (αGal) carbohydrate antigen. This situation effectively eliminates any role for anti-Gal antibody in GTKO cardiac xenograft rejection. Survival of heterotopic GTKO cardiac xenografts in nonhuman primates continues to increase. GTKO graft rejection commonly involves vascular antibody deposition and variable complement deposition. Non-Gal antibody responses to porcine antigens associated with inflammation, complement, and hemostatic regulation and to new carbohydrate antigens have been identified. Their contribution to rejection remains under investigation. Orthotopic CXTx is limited by early perioperative cardiac xenograft dysfunction (PCXD). However, hearts affected by PCXD recover full cardiac function and orthotopic survival up to 2 months without rejection has been reported. SUMMARY: CXTx remains a promising technology for treating end-stage cardiac failure. Genetic modification of the donor and refinement of immunosuppressive regimens have extended heterotopic cardiac xenograft survival from minutes to in excess of 8 months.


Subject(s)
Graft Rejection/immunology , Heart Failure/surgery , Heart Transplantation , Transplantation, Heterologous/immunology , Animals , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Expression , Primates , Swine , Thrombosis/genetics , Thrombosis/immunology , Tissue Donors/supply & distribution , Transplants
16.
Transplantation ; 91(9): 968-75, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21403591

ABSTRACT

BACKGROUND: This study compares the pathologic condition of delayed xenograft rejection in Gal-positive and Gal-knockout cardiac xenografts after pig-to-baboon heterotopic cardiac xenotransplantation when the induced anti-Gal antibody response is unregulated, blocked, or absent. METHODS: Baboon recipients of Gal-positive, CD46 pig hearts were treated with an αGal polymer (group 1; n=11) or Gal-specific immunoapheresis (group 2; n=8) to block anti-Gal antibody. Gal-knockout cardiac xenografts recipients (group 3; n=5) received no anti-Gal therapy. Perioperative and interim biopsies were examined and antibody responses were determined. RESULTS: No hyperacute rejection was seen and histologic findings were similar across the groups. All groups showed vascular antibody deposition in perioperative and interim biopsies and in explant samples. A prominent antibody response was detected only in group 2. Complement activation was evident by C3d deposition but deposition of C5b and C5b-9 was limited. Earliest evidence of myocardial injury was myocyte vacuolization in the absence of microvascular thrombosis or coagulative necrosis that developed later. Histology of explanted hearts exhibited mainly microvascular thrombosis and coagulative necrosis with little evidence of interstitial hemorrhage or edema. CONCLUSIONS: The histology of rejection seemed independent of the anti-Gal or non-Gal immune response. Myocyte vacuolization seems to be an early feature of delayed xenograft rejection presaging more classic pathologic features.


Subject(s)
Graft Rejection/etiology , Graft Rejection/immunology , Heart Transplantation/adverse effects , Heart Transplantation/immunology , Trisaccharides/immunology , Animals , Antibodies, Heterophile/blood , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Knockout Techniques , Graft Rejection/enzymology , Graft Rejection/pathology , Heart Transplantation/pathology , Microvessels/pathology , Myocytes, Cardiac/pathology , Necrosis , Papio , Swine , Thrombosis/pathology , Transplantation, Heterologous
17.
Xenotransplantation ; 18(1): 14-27, 2011.
Article in English | MEDLINE | ID: mdl-21342284

ABSTRACT

BACKGROUND: Gene profiling methods have been widely useful for delineating changes in gene expression as an approach for gaining insight into the mechanism of rejection or disease pathology. Herein, we use gene profiling to compare changes in gene expression associated with different orthotopic cardiac xenotransplantation (OCXTx) outcomes and to identify potential effects of OCXTx on cardiac physiology. METHODS: We used the Affymetrix GeneChip Porcine Genomic Array to characterize three types of orthotopic cardiac xenograft outcomes: 1) rejected hearts that underwent delayed xenograft rejection (DXR); 2) survivor hearts in which the xenograft was not rejected and recipient death was due to model complications; and 3) hearts which failed to provide sufficient circulatory support within the first 48 h of transplant, termed "perioperative cardiac xenograft dysfunction" (PCXD). Gene expression in each group was compared to control, not transplanted pig hearts, and changes in gene expression > 3 standard deviations (±3SD) from the control samples were analyzed. A bioinformatics analysis was used to identify enrichments in genes involved in Kyoto Encyclopedia of Genes and Genomes pathways and gene ontogeny molecular functions. Changes in gene expression were confirmed by quantitative RT-PCR. RESULTS: The ±3SD data set contained 260 probes, which minimally exhibited a 3.5-fold change in gene expression compared to control pig hearts. Hierarchical cluster analysis segregated rejected, survivor and PCXD samples, indicating a unique change in gene expression for each group. All transplant outcomes shared a set of 21 probes with similarly altered expression, which were indicative of ongoing myocardial inflammation and injury. Some outcome-specific changes in gene expression were identified. Bioinformatics analysis detected an enrichment of genes involved in protein, carbohydrate and branched amino acid metabolism, extracellular matrix-receptor interactions, focal adhesion, and cell communication. CONCLUSIONS: This is the first genome wide assessment of changes in cardiac gene expression after OCXTx. Hierarchical cluster analysis indicates a unique gene profile for each transplant outcome but additional samples will be required to define the unique classifier probe sets. Quantitative RT-PCR confirmed that all transplants exhibited strong evidence of ongoing inflammation and myocardial injury consistent with the effects of cytokines and vascular antibody-mediated inflammation. This was also consistent with bioinformatic analysis suggesting ongoing tissue repair in survivor and PCXD samples. Bioinformatics analysis suggests for the first time that xenotransplantation may affect cardiac metabolism in survivor and rejected samples. This study highlights the potential utility of molecular analysis to monitor xenograft function, to identify new molecular markers and to understand processes, which may contribute to DXR.


Subject(s)
Gene Expression , Heart Transplantation/methods , Heart/physiology , Transplantation, Heterologous/methods , Animals , Cluster Analysis , Computational Biology , Gene Expression Profiling , Graft Rejection/pathology , Humans , Oligonucleotide Array Sequence Analysis , Sus scrofa
18.
Xenotransplantation ; 18(1): 6-13, 2011.
Article in English | MEDLINE | ID: mdl-21342283

ABSTRACT

BACKGROUND: After substantial progress on many fronts, one of the remaining barriers still opposing the clinical application of xenotransplantation is a disseminated intravascular coagulopathy (DIC) that is observed in the pre-clinical model of porcine-to-primate transplantation. The onset of DIC is particularly rapid in recipients of pulmonary xenografts, usually occurring within the first days or even hours of reperfusion. METHODS: In this study, we describe the results of two porcine-to-baboon transplants utilizing porcine lungs depleted of macrophages, deficient in the α-1,3-galactosyltransferase gene, and with the expression of human decay-accelerating factor, a complement regulatory protein. RESULTS: In both cases, evidence of DIC was observed within 48 h of reperfusion, with thrombocytopenia and increases in levels of thrombin-antithrombin complex evident in both cases. Depletion of fibrinogen was observed in one graft, whereas elevation of D-dimer levels was observed in the other. One graft, which showed focal lymphocytic infiltrates pre-operatively, failed within 3 h. CONCLUSIONS: The results indicate that further efforts to address the coagulopathy associated with pulmonary xenotransplantation are needed. Further, evidence suggests that resident porcine immune cells can play an important role in the coagulopathy associated with xenotransplantation.


Subject(s)
Blood Coagulation Disorders/immunology , Galactosyltransferases/genetics , Lung Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Antithrombins/metabolism , CD55 Antigens/genetics , CD55 Antigens/immunology , Fibrin Fibrinogen Degradation Products/metabolism , Galactosyltransferases/metabolism , Gene Knockdown Techniques , Graft Survival , Humans , Papio/immunology , Swine/immunology , Thrombin/metabolism , Transplantation, Heterologous/pathology
19.
J Thorac Cardiovasc Surg ; 141(1): 269-75, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21168032

ABSTRACT

OBJECTIVES: Human subjects and Old World primates have high levels of antibody to galactose-α-1,3 galactose ß-1,4-N-acetylglucosamine (α-Gal). Commercially available bioprosthetic heart valves of porcine and bovine origin retain the Gal antigen despite current processing techniques. Gal-deficient pigs eliminate this xenoantigen. This study tests whether binding of human anti-Gal antibody effects calcification of wild-type and Gal-deficient glutaraldehyde-fixed porcine pericardium by using a standard subcutaneous implant model. METHODS: Expression of α-Gal was characterized by lectin Griffonia simplicifolia-IB4 staining. Glutaraldehyde-fixed pericardial disks from Gal-positive and Gal-deficient pigs were implanted into 12-day-old Wistar rats and 1.5-kg rabbits with and without prelabeling with affinity-purified human anti-Gal antibody. Calcification of the implants was determined after 3 weeks by using inductively coupled plasma spectroscopy. RESULTS: The α-Gal antigen was detected in wild-type but not Gal-deficient porcine pericardium. Wild-type disks prelabeled with human anti-Gal antibody exhibited significantly greater calcification compared with that seen in antibody-free wild-type samples (mean ± standard error of the mean: 111 ± 8.4 and 74 ± 9.6 mg/g, respectively; P = .01). In the presence of anti-Gal antibody, a significantly greater level of calcification was detected in wild-type compared with GTKO porcine pericardium (111 ± 8.4 and 55 ± 11.8 mg/g, respectively; P = .005). Calcification of Gal-deficient pericardium was not affected by the presence of anti-Gal antibody (51 ± 9.1 and 55 ± 11.8 mg/g). CONCLUSIONS: In this model anti-Gal antibody accelerates calcification of wild-type but not Gal-deficient glutaraldehyde-fixed pericardium. This study suggests that preformed anti-Gal antibody present in all patients might contribute to calcification of currently used bioprosthetic heart valves. Gal-deficient pigs might become the preferred source for new, potentially calcium-resistant bioprosthetic heart valves.


Subject(s)
Antigens, Heterophile/immunology , Bioprosthesis , Calcinosis/immunology , Heart Valve Prosthesis Implantation/instrumentation , Heart Valve Prosthesis , Pericardium/transplantation , Trisaccharides/immunology , Animals , Animals, Genetically Modified , Antigens, Heterophile/biosynthesis , Autoantibodies/administration & dosage , Fixatives , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Glutaral , Heart Valve Prosthesis Implantation/adverse effects , Humans , Microscopy, Fluorescence , Pericardium/immunology , Plant Lectins , Prosthesis Design , Rabbits , Rats , Rats, Wistar , Swine/genetics , Time Factors , Transplantation, Heterologous , Trisaccharides/biosynthesis
20.
Transplantation ; 91(3): 287-92, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21119562

ABSTRACT

BACKGROUND: α1,3-Galactosyltransferase gene knockout (GTKO) pigs reduced the significance of antibody to galactose alpha 1,3-galactose (Gal) antigens but did not eliminate delayed xenograft rejection (DXR). We hypothesize that DXR of GTKO organs results from an antibody response to a limited number of non-Gal endothelial cell (EC) membrane antigens. In this study, we screened a retrovirus expression library to identify EC membrane antigens detected after cardiac xenotransplantation. METHODS: Expression libraries were made from GT:CD46 and GTKO porcine aortic ECs. Viral stocks were used to infect human embryonic kidney cells (HEK) that were selected by flow cytometry for IgG binding from sensitized cardiac heterotopic xenograft recipients. After three to seven rounds of selection, individual clones were assessed for non-Gal IgG binding. The porcine complementary DNA was recovered by polymerase chain reaction amplification, sequenced, and identified by homology comparisons. RESULTS: A total of 199 and 317 clones were analyzed from GT:CD46 and GTKO porcine aortic EC complementary DNA libraries, respectively. Sequence analysis identified porcine CD9, CD46, CD59, and the EC protein C receptor. We also identified porcine annexin A2 and a glycosyltransferase with homology to the human ß1,4 N-acetylgalactosaminyl transferase 2 gene. CONCLUSION: The identified proteins include key EC functions and suggest that non-Gal antibody responses may compromise EC functions and thereby contribute to DXR. Recovery of the porcine ß1,4 N-acetylgalactosaminyl transferase 2 suggests that an antibody response to a SD-like carbohydrate may represent a new carbohydrate moiety involved in xenotransplantation. The identification of these porcine gene products may lead to further donor modification to enhance resistance to DXR and further reduce the level of xenograft antigenicity.


Subject(s)
Antigens/metabolism , Carbohydrates/immunology , Heart Transplantation/immunology , Membrane Proteins/immunology , Transplantation, Heterologous/immunology , Animals , Endothelium, Vascular/immunology , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Gene Knockout Techniques , Graft Rejection/immunology , Humans , Models, Animal , Primates , Retroviridae , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...