Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.557
Filter
1.
Zoolog Sci ; 30(7): 577-84, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23829218

ABSTRACT

Urodele amphibians are thought to have poorer immune responses than evolutionary more ancestral vertebrate classes, such as bony fish. We investigated skin graft rejection and transplantation immunity in Urodele amphibians, Japanese newts, and Asiatic salamanders, and compared these findings to those from transplants in several species of frogs. The skin grafts used in this study were either allogeneic or xenogeneic. The mean survival time of the first set of allografts at 20°C was approximately 60 days for chronic responses in Urodela and 20 days for acute responses in Anura. As the graft survival times of urodeles were significantly longer than those of anurans, even when urodeles were repeatedly grafted from identical donors, there appear to be substantial differences in transplantation immunity between Urodela and Anura. These slow responses in Urodela may not be accompanied by the expansion of cytotoxic T cells, as observed in fish and anuran species, which are known to have functional major histocompatibility complex (MHC)-class I systems. In our study, approximately five histo-incompatible immunogenic components were found to be involved in chronic responses in newts. Similar chronic responses were also observed in xenograft rejection in newts. In contrast, xenografts were rejected in frogs due to an accelerated acute response, possibly involving natural killer cells. Our findings that some anti-allogeneic components appear to be shared with xenogeneic components indicate that the diversification of the acquired immune system is poorly developed in Urodela.


Subject(s)
Graft Rejection/immunology , Skin Transplantation/immunology , Transplantation, Heterologous/immunology , Urodela/immunology , Animals , Wound Healing/immunology
2.
J Immunol ; 191(5): 2796-805, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23872050

ABSTRACT

Significant immunological obstacles are to be negotiated before xenotransplantation becomes a clinical reality. An initial rejection of transplanted vascularized xenograft is attributed to Galα1,3Galß1,4GlcNAc-R (Galα1,3-Gal)-dependent and -independent mechanisms. Hitherto, no receptor molecule has been identified that could account for Galα1,3-Gal-independent rejection. In this study, we identify the tetraspanin CD82 as a receptor molecule for the Galα1,3-Gal-independent mechanism. We demonstrate that, in contrast to human undifferentiated myeloid cell lines, differentiated cell lines are capable of recognizing xenogeneic porcine aortic endothelial cells in a calcium-dependent manner. Transcriptome-wide analysis to identify the differentially expressed transcripts in these cells revealed that the most likely candidate of the Galα1,3-Gal-independent recognition moiety is the tetraspanin CD82. Abs to CD82 inhibited the calcium response and the subsequent activation invoked by xenogeneic encounter. Our data identify CD82 on innate immune cells as a major "xenogenicity sensor" and open new avenues of intervention to making xenotransplantation a clinical reality.


Subject(s)
Graft Rejection/immunology , Kangai-1 Protein/immunology , Transplantation, Heterologous/immunology , Animals , Blotting, Western , Endothelial Cells/immunology , Flow Cytometry , Fluorescent Antibody Technique , Humans , Microscopy, Confocal , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Swine
4.
Clin Exp Immunol ; 172(2): 349-62, 2013 May.
Article in English | MEDLINE | ID: mdl-23574330

ABSTRACT

Oxazolone-induced colitis in mice has become a recognized model to study the efficacy of therapeutics targeting the immunological response underlying the development of inflammatory bowel disease. However, this model cannot be used when therapeutics designed to address human targets do not interact with the respective murine counterpart. In this study, we examined the induction of oxazolone mediated colitis in non-obese diabetic-severe combined immunodeficiency interleukin-2Rγ(null) (NOD-SCID IL2Rγ(null)) mice engrafted with human peripheral blood mononuclear cells (hPBMC) derived from patients suffering from ulcerative colitis (UC), atopic dermatitis (AD) and healthy volunteers. NOD-SCID IL2Rγ (null) mice were engrafted with hPBMC followed by challenge with oxazolone or ethanol vehicle. Mice developed the same symptoms as observed previously in immunocompetent mice. The clinical activity score increased and the colon architecture was characterized by the development of oedema, fibrosis, crypt loss and dense infiltration of predominantly T cells into the lamina propria. Fluorescence activated cell sorter (FACS) analysis of lymphocytes in the colon identified natural killer (NK) T cells as a major constituent. In contrast to studies with immunocompetent mice, we observed the same phenotype in the group challenged with ethanol vehicle. The phenotype was most pronounced in mice engrafted with PBMC derived from a patient suffering from UC, suggesting that the immunological history of the donors predisposes the engrafted mice to react to ethanol. The model described here has the potential to study the efficacy of therapeutics targeting human lymphocytes in a model which is more reflective of the human disease. In addition, it might be developed to elucidate molecular mechanisms underlying the disease.


Subject(s)
Colitis, Ulcerative/immunology , Dermatitis, Atopic/immunology , Ethanol/pharmacology , Leukocytes, Mononuclear/transplantation , Oxazolone/pharmacology , Animals , Cell Line , Colitis, Ulcerative/chemically induced , Disease Models, Animal , Graft vs Host Disease/immunology , Humans , Immunoglobulin E/blood , Immunoglobulin G/blood , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Receptors, Interleukin-2/genetics , Transplantation, Heterologous/immunology
5.
PLoS One ; 8(4): e60680, 2013.
Article in English | MEDLINE | ID: mdl-23585844

ABSTRACT

Human cells from acute myeloid leukemia (AML) patients are frequently transplanted into immune-compromised mouse strains to provide an in vivo environment for studies on the biology of the disease. Since frequencies of leukemia re-initiating cells are low and a unique cell surface phenotype that includes all tumor re-initiating activity remains unknown, the underlying mechanisms leading to limitations in the xenotransplantation assay need to be understood and overcome to obtain robust engraftment of AML-containing samples. We report here that in the NSG xenotransplantation assay, the large majority of mononucleated cells from patients with AML fail to establish a reproducible myeloid engraftment despite high donor chimerism. Instead, donor-derived cells mainly consist of polyclonal disease-unrelated expanded co-transplanted human T lymphocytes that induce xenogeneic graft versus host disease and mask the engraftment of human AML in mice. Engraftment of mainly myeloid cell types can be enforced by the prevention of T cell expansion through the depletion of lymphocytes from the graft prior transplantation.


Subject(s)
Bone Marrow Transplantation/pathology , Graft vs Host Disease/pathology , Leukemia, Myeloid, Acute/pathology , Myeloid Cells/pathology , T-Lymphocytes/pathology , Transplantation, Heterologous/pathology , Adult , Aged , Animals , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/mortality , Cell Proliferation , Female , Graft vs Host Disease/immunology , Humans , Leukemia, Myeloid, Acute/immunology , Lymphocyte Depletion , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Myeloid Cells/immunology , Neoplasm Transplantation , Survival Analysis , T-Lymphocytes/immunology , Transplantation Chimera , Transplantation, Heterologous/immunology , Transplantation, Heterologous/mortality , Whole-Body Irradiation
6.
Xenotransplantation ; 20(2): 110-22, 2013.
Article in English | MEDLINE | ID: mdl-23489741

ABSTRACT

BACKGROUND: Stem cell therapy may help restore cardiac function after acute myocardial infarction (AMI), but the optimal therapeutic cell type has not been identified. METHODS: We examined the effects of CD34-/CD45- human unrestricted somatic stem cells (USSCs) in pigs (n = 30) with an AMI created by a 90-min occlusion of the left anterior descending coronary artery. Pigs were randomly assigned to receive either USSCs (302 ± 23 × 10(6) cells) or phosphate-buffered saline via 15 NOGA-guided transendocardial injections 10 days after AMI. Cyclosporine A (10 mg/kg orally, twice a day) was started in all pigs 3 days before control or cell treatment. Cardiac function was assessed by echocardiography before injection and at 4 and 8 weeks after treatment. Serum titers for pig IgG antibodies against USSCs were also measured at these time points and before AMI. RESULTS: Compared with control pigs, USSC-treated pigs showed no significant differences in any of the functional parameters examined. USSC-treated pigs showed variable increases in anti-USSC IgG antibody titers in the blood and chronic inflammatory infiltrates at the cell injection sites. Immunohistochemical studies of the injection sites using human anti-mitochondrial antibodies failed to detect implanted USSCs. CONCLUSIONS: We conclude that human USSCs did not improve cardiac function in a pig model of AMI. Cell transplantation in a xenogeneic setting may obscure the benefits of stem cell therapy.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Myocardial Infarction/therapy , Pluripotent Stem Cells/transplantation , Transplantation, Heterologous/methods , Animals , Antibodies, Heterophile/blood , Antibodies, Heterophile/immunology , Antigens, Heterophile/immunology , Cells, Cultured , Disease Models, Animal , Heart Function Tests , Humans , Immunosuppressive Agents/therapeutic use , Mitochondria/immunology , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Pluripotent Stem Cells/cytology , Random Allocation , Sus scrofa , Transplantation, Heterologous/immunology , Treatment Failure
7.
Xenotransplantation ; 20(2): 123-30, 2013.
Article in English | MEDLINE | ID: mdl-23489828

ABSTRACT

BACKGROUND: Delayed xenograft rejection (DXR) involves type II vascular endothelial cell (VEC) activation including upregulation of pro-inflammatory genes, which contributes to infiltration into the graft and a complex process of cytokine production. Approaches to prevent DXR have shown limited success. In this study, we modified heart donors using siRNA in an attempt to attenuate DXR and to improve xenograft survival in the mouse-to-rat heterotopic heart transplant model. METHODS: siRNA technology was used to inhibit NF-kappaB p65 gene expression in vivo in mice. After the donor was transfected with siRNA, the effects of NF-kappaB siRNA on DXR and expression of NF-kappaB and pro-inflammatory genes were evaluated in a concordant mouse-to-rat cardiac xenograft model. RESULTS: Treatment of NF-kappaB siRNA prolonged median heart graft survival time in the recipient rats from 1.7 days in a PBS control group to 5.4 days in the NF-kappaB siRNA-treated group (P < 0.05). Compared with normal mouse hearts, the NF-kappaB p65 mRNA relative levels following siRNA injection in the donors decreased significantly (approximately 70% reduction) in grafts harvested 12 h after transplantation. The mRNA levels of VCAM-1, ICAM-1, and interleukin-1 displayed a similar reduction. Histological evaluation using light and electron microscopy showed that damage of endothelial cells after NF-kappaB siRNA treament occured at a later time. CONCLUSION: Transfection of NF-kappaB p65 siRNA in donor animals can delay the emergence of DXR. This treatment may be used as part of strategies to minimize the complex and multi-faceted rejection responses in vascularized xenografts.


Subject(s)
Genetic Therapy/methods , Graft Rejection/immunology , Graft Rejection/therapy , Heart Transplantation/immunology , Transcription Factor RelA/genetics , Transplantation, Heterologous/immunology , Animals , Graft Rejection/pathology , Graft Survival/immunology , Heart Transplantation/adverse effects , Humans , Intercellular Adhesion Molecule-1/genetics , Interleukin-1/genetics , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Transcription Factor RelA/immunology , Transfection , Transplantation, Heterologous/adverse effects , Vascular Cell Adhesion Molecule-1/genetics
8.
Xenotransplantation ; 20(1): 27-35, 2013.
Article in English | MEDLINE | ID: mdl-23384142

ABSTRACT

BACKGROUND: Clinical xenotransplantation is not possible because humans possess antibodies that recognize antigens on the surface of pig cells. Galα-1,3-Gal (Gal) and N-glycolylneuraminic acid (Neu5Gc) are two known xenoantigens. METHODS: We report the homozygous disruption of the α1, 3-galactosyltransferase (GGTA1) and the cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH) genes in liver-derived female pig cells using zinc-finger nucleases (ZFNs). Somatic cell nuclear transfer (SCNT) was used to produce healthy cloned piglets from the genetically modified liver cells. Antibody-binding and antibody-mediated complement-dependent cytotoxicity assays were used to examine the immunoreactivity of pig cells deficient in Neu5Gc and Gal. RESULTS: This approach enabled rapid production of a pig strain deficient in multiple genes without extensive breeding protocols. Immune recognition studies showed that pigs lacking both CMAH and GGTA1 gene activities reduce the humoral barrier to xenotransplantation, further than pigs lacking only GGTA1. CONCLUSIONS: This technology will accelerate the development of pigs for xenotransplantation research.


Subject(s)
Disaccharides/immunology , Neuraminic Acids/immunology , Sus scrofa/genetics , Sus scrofa/immunology , Transplantation, Heterologous/immunology , Animals , Antibodies, Heterophile/metabolism , Antibody-Dependent Cell Cytotoxicity , Antigens, Heterophile/immunology , Antigens, Heterophile/metabolism , Base Sequence , Cells, Cultured , DNA/genetics , Disaccharides/deficiency , Female , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Gene Knockout Techniques/methods , Humans , Leukocytes, Mononuclear/immunology , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/genetics , Neuraminic Acids/metabolism , Sus scrofa/metabolism
10.
Xenotransplantation ; 20(2): 100-9, 2013.
Article in English | MEDLINE | ID: mdl-23442186

ABSTRACT

BACKGROUND: We investigated whether graft produced anti-human CD2, mediated by adenovirus (Adv) transduction of pig neonatal islet cell clusters (pNICC), would protect xenografts in a humanized mouse model from immune attack and whether such immunosuppression would remain local. METHODS: A mouse anti-human CD2 Ab (CD2hb11) previously generated by us was genetically engineered to produce chimeric and humanized versions. The three forms of CD2hb11 were named dilimomab (mouse), diliximab (chimeric) and dilizumab (humanized). All 3 forms of CD2hb11 Ab were tested for their ability to bind CD3(+) human T cells and to inhibit a human anti-pig xenogeneic mixed lymphocyte reaction (MLR). They were administered systemically in a humanized mouse model in order to test their ability to deplete human CD3(+) T cells and whether they induced a cytokine storm. An adenoviral vector expressing diliximab was generated for transduction of pNICC. Humanized mice were transplanted with either control-transduced pNICC or diliximab-transduced pNICC and human T cells within grafts and spleens were enumerated by flow cytometry. RESULTS: Dilimomab and diliximab inhibited a human anti-pig xenogeneic response but dilizumab did not. All 3 forms of CD2hb11 Ab bound human T cells in vitro though dilimomab and diliximab exhibited 300-fold higher avidity than dilizumab. All 3 anti-CD2 Abs could deplete human CD3(+) T cells in vivo in a humanized mouse model without inducing upregulation of activation markers or significant release of cytokines. Humanized mice transplanted with diliximab-transduced pNICC afforded depletion of CD3(+) T cells at the graft site leaving the peripheral immune system intact. CONCLUSIONS: Local production of a single Ab against T cells can reduce graft infiltration at the xenograft site and may reduce the need for conventional, systemic immunosuppression.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD2 Antigens/immunology , Immunosuppressive Agents/pharmacology , Islets of Langerhans Transplantation/immunology , T-Lymphocytes/immunology , Transplantation, Heterologous/immunology , Adenoviridae/genetics , Animals , Antibodies, Heterophile/immunology , Antibodies, Heterophile/pharmacology , Antibodies, Monoclonal/immunology , Antigens, Heterophile/genetics , Antigens, Heterophile/immunology , CD2 Antigens/genetics , Chimera , Flow Cytometry , Graft Rejection/immunology , Humans , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred NOD , Mice, SCID , Species Specificity
11.
Xenotransplantation ; 20(1): 18-26, 2013.
Article in English | MEDLINE | ID: mdl-23347130

ABSTRACT

BACKGROUND: Endothelial damage is a critical step in the development of (xeno) transplantation-related and cardiovascular pathology. In humans, the amount of circulating endothelial cells (CEC) correlates to disease intensity and functions as a valuable damage marker. While (xeno) transplantation and cardiovascular research is regularly performed in porcine models, the paucity of antibodies against porcine endothelium epitopes hinders the use of CEC as damage marker. OBJECTIVE: This study aimed to develop a method for porcine CEC detection using anti-human antibodies against porcine endothelium epitopes. METHODS: Human umbilical vein endothelial cells (HUVEC, control) and their swine equivalent (SUVEC) were used to assess the cross-species immunoreactivity of fluorescently labeled anti-human CD31/CD51/CD54/CD62E/CD105/CD106/CD144/CD146/PAL-E/lectin-1/vWF antibodies by isotype-controlled fluorescence-activated cell sorting (FACS) and confocal microscopy. Next, reactivity was ascertained with mature porcine kidney-derived endothelial cells (PKEC), and a FACS-based whole blood CEC quantification method was employed using osmotic erythrolysis and CD105 and CD146 double staining after CD45 exclusion. RESULTS: Of the 21 assayed antibodies, the MEM-229 clone of CD105 and P1H12 clone of CD146 showed immunoreactivity with SUVEC and PKEC. Double staining showed baseline porcine CEC count of 673.1 ± 551.4 CEC/ml, while the first 7.5 ml of drawn blood (representative of vascular damage) contained 1118 ± 661.4 CEC/ml (n = 14, P = 0.04). A second experiment (n = 5) including CD45 exclusion identified only 14.5 ± 10.8% double-positive CD105-146 events per ml blood. CONCLUSION: Porcine endothelium can be specifically labeled using anti-human CD146 and CD105 antibodies. These antibodies can therefore be used for the identification and quantification of CEC in porcine whole blood by FACS after osmotic erythrolysis.


Subject(s)
Blood Cells/cytology , Blood Cells/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Sus scrofa/blood , Sus scrofa/immunology , Animals , Antibodies, Heterophile/immunology , Antigens, CD/immunology , Antigens, Heterophile/immunology , CD146 Antigen/immunology , Cell Count/methods , Cell Count/veterinary , Cells, Cultured , Cross Reactions , Endoglin , Flow Cytometry , Human Umbilical Vein Endothelial Cells , Humans , Microscopy, Confocal , Receptors, Cell Surface/immunology , Transplantation, Heterologous/adverse effects , Transplantation, Heterologous/immunology
12.
Blood ; 121(8): 1316-25, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23293079

ABSTRACT

Current mouse lines efficient for human cell xenotransplantation are backcrossed into NOD mice to introduce its multiple immunodeficient phenotypes. Our positional genetic study has located the NOD-specific polymorphic Sirpa as a molecule responsible for its high xenograft efficiency: it recognizes human CD47 and the resultant signaling may cause NOD macrophages not to engulf human grafts. In the present study, we established C57BL/6.Rag2(nullIl2rgnull) mice harboring NOD-Sirpa (BRGS). BRGS mice engrafted human hematopoiesis with an efficiency that was equal to or even better than that of the NOD.Rag1(nullIl2rgnull) strain, one of the best xenograft models. Consequently, BRGS mice are free from other NOD-related abnormalities; for example, they have normalized C5 function that enables the evaluation of complement-dependent cytotoxicity of antibodies against human grafts in the humanized mouse model. Our data show that efficient human cell engraftment found in NOD-based models is mounted solely by their polymorphic Sirpa. The simplified BRGS line should be very useful in future studies of human stem cell biology.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Receptors, Immunologic/genetics , Transplantation, Heterologous/immunology , Transplantation, Heterologous/methods , Animals , Antibodies, Heterophile/immunology , CD47 Antigen/immunology , CD47 Antigen/metabolism , Cell Lineage/immunology , DNA-Binding Proteins/genetics , Female , Graft Survival/immunology , Hematopoiesis/physiology , Humans , Lymphocytes/cytology , Lymphocytes/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Models, Animal , Phagocytosis/immunology , Polymorphism, Genetic/immunology , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction/immunology
13.
Cytokine ; 61(1): 118-26, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23058476

ABSTRACT

IL-15 is a pleotropic cytokine, which plays an important role in natural killer (NK) cell activity, T cell proliferation, and T cell cytotoxic activity. Dendritic cells (DCs) are the major antigen presenting cells in the immune system and presumed to play an important role in immune recognition of allo and xenotransplantation. We showed that IL-15 activated human peripheral blood DC is cytotoxic to human and porcine aortic endothelial cells. Unlike DCs, CD14+ monocytes show no cytotoxicity against the endothelial cells. This cytotoxic potential of IL-15 activated DC against endothelial cells is dose dependent and increases significantly upon treatment of endothelial cells with inflammatory cytokines like TNF-α or IFN-γ. The cytotoxic potential of IL-15 activated DC is associated with apoptosis of endothelial cells, as indicated by the increased Annexin V staining, caspase activation and loss of mitochondrial membrane potential. Further it was observed that DC mediated cytotoxicity against endothelial cell is mediated via granzyme B possibly secreted by the activated DCs.


Subject(s)
Apoptosis/immunology , Dendritic Cells/immunology , Endothelial Cells/immunology , Interleukin-15/immunology , Animals , Aorta/immunology , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Granzymes/metabolism , Humans , Interferon-gamma/pharmacology , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Lipopolysaccharide Receptors , Lymphocyte Activation/immunology , Membrane Potential, Mitochondrial , Monocytes/metabolism , Swine , T-Lymphocytes, Cytotoxic/immunology , Transplantation, Heterologous/immunology , Transplantation, Homologous/immunology , Tumor Necrosis Factor-alpha/pharmacology
14.
Xenotransplantation ; 20(1): 5-17, 2013.
Article in English | MEDLINE | ID: mdl-23190260

ABSTRACT

BACKGROUND: Various groups have reported prolonged diabetes reversal and graft function after porcine islet transplantation into diabetic macaques using different experimental designs (macaque source, islet source, type of immunosuppression): subsequently, the International Xenotransplantation Association has published recommendations for entering a clinical trial. Our experiments showed limitations that affected consistent achievement of long-term survival. We aimed to identify these limitations and underlying causes to emphasize the translational value of this highly relevant type 1 diabetic macaque model. METHODS: We reviewed data from our institution and literature data on long-term porcine islet xenograft survival in the diabetic macaque model, especially focusing on aspects of incomplete diabetes reversal relative to macaque normal values. This phenomenon was compared with diabetes reversal in an allo-islet transplant model in macaques and with chronic insulin treatment of diabetic macaques, all with 180-day follow-up. This comparison enabled to identify potential model limitations and underlying causative factors. RESULTS: Especially in the xenograft model, the achievement of long-term graft survival revealed limitations including chronic, mild hyperglycemia and absence of body weight (BW) gain or even progressive BW loss. Metabolic incompatibilities in glycemic control (i.e., insulin kinetics) between the pig and macaque species underlie chronic, mild hyperglycemia. This phenomenon might not bear relevance for the pig-to-human species combination because the glycemic control in pigs and humans is similar and differs from that in nonhuman primates (NHP). Weight loss could be related to changes in the gastrointestinal tract related with local high exposure to orally administered immunosuppressants; these must be given at higher dose levels because of low bioavailability in macaques to achieve systemic exposure at therapeutic levels. This is aggravated by insufficient graft insulin production in proportion to the needs of macaques: this model limitation has no translational value to the pig-to-human setting. Nutritional deficits can result in incorrect interpretation of blood glucose levels and C-peptide levels regarding graft function. Likewise, nutritional status alters physiologic responses, influencing susceptibility to infectious and noninfectious complications. CONCLUSION: THE model-induced confounding described interferes with accurate interpretation of safety and efficacy studies, which affects the translational value of pig-to-NHP islet cell transplant studies to the pig-to-human transplant condition.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/methods , Transplantation, Heterologous , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Graft Survival , Humans , Immunosuppression Therapy , Islets of Langerhans Transplantation/adverse effects , Islets of Langerhans Transplantation/immunology , Macaca , Models, Animal , Sus scrofa , Transplantation, Heterologous/adverse effects , Transplantation, Heterologous/immunology , Weight Loss
15.
J Biosci Bioeng ; 115(2): 207-15, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23068617

ABSTRACT

Decellularized biologic matrices are plausible biomedical materials for the bioengineering in liver transplantation. However, one of the concerns for safe medical application is the lack of objective assessment of the immunogen within the materials and the in vivo immune responses to the matrices. The purpose of this study was the production of immunogen-reduced and biocompatible matrices from porcine liver. In the present study, 0.1% SDS solution was effective for removing DNA fragments and sequences encoding possible immunogenic and viral antigens within the matrices. The PCR analysis showed that galactose-α-1,3 galactose ß-1,4-N-acetylglucosamine (1,3 gal), swine leukocyte antigen (SLA), and porcine endogenous retrovirus (PERV) were completely removed in the matrices. Collagen and glycosaminoglycans (GAGs) were preserved over 63%-71%, respectively, compared to those of native liver. The implanted decellularized tissues showed minimal host responses and naturally degraded within 10 weeks. In this study, we produced immunogen-reduced and biocompatible extracellular matrices from porcine liver. Although future investigations would be required to determine the mechanism of the host reaction, this study could provide useful information of porcine liver-derived biologic matrices for liver researches.


Subject(s)
Biocompatible Materials , Extracellular Matrix/immunology , Liver Transplantation/methods , Liver/cytology , Liver/immunology , Sus scrofa , Tissue Scaffolds , Transplantation, Heterologous/methods , Animals , Antigens/analysis , Antigens/genetics , Antigens/immunology , Antigens/isolation & purification , Cell Separation , Collagen/analysis , DNA/analysis , DNA/genetics , Endogenous Retroviruses/immunology , Endogenous Retroviruses/isolation & purification , Extracellular Matrix/metabolism , Glycosaminoglycans/analysis , Graft Rejection/immunology , Guided Tissue Regeneration/methods , Histocompatibility Antigens Class I , Histocompatibility Antigens Class II/analysis , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/isolation & purification , Liver Transplantation/immunology , Sus scrofa/immunology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Transplantation, Heterologous/immunology
17.
Xenotransplantation ; 19(6): 342-54, 2012.
Article in English | MEDLINE | ID: mdl-23145497

ABSTRACT

BACKGROUND: The increasing availability of genetically engineered pigs is steadily improving the results of pig organ and cell transplantation in non-human primates (NHPs). Current techniques offer knockout of pig genes and/or knockin of human genes. Knowledge of normal values of hematologic, biochemical, coagulation, and other parameters in healthy genetically engineered pigs and NHPs is important, particularly following pig organ transplantation in NHPs. Furthermore, information on parameters in various NHP species may prove important in selecting the optimal NHP model for specific studies. METHODS: We have collected hematologic, biochemical, and coagulation data on 71 α1,3-galactosyltransferase gene-knockout (GTKO) pigs, 18 GTKO pigs additionally transgenic for human CD46 (GTKO.hCD46), four GTKO.hCD46 pigs additionally transgenic for human CD55 (GTKO.hCD46.hCD55), and two GTKO.hCD46 pigs additionally transgenic for human thrombomodulin (GTKO.hCD46.hTBM). RESULTS: We report these data and compare them with similar data from wild-type pigs and the three major NHP species commonly used in biomedical research (baboons, cynomolgus, and rhesus monkeys) and humans, largely from previously published reports. CONCLUSIONS: Genetic modification of the pig (e.g., deletion of the Gal antigen and/or the addition of a human transgene) (i) does not result in abnormalities in hematologic, biochemical, or coagulation parameters that might impact animal welfare, (ii) seems not to alter metabolic function of vital organs, although this needs to be confirmed after their xenotransplantation, and (iii) possibly (though, by no means certainly) modifies the hematologic, biochemical, and coagulation parameters closer to human values. This study may provide a good reference for those working with genetically engineered pigs in xenotransplantation research and eventually in clinical xenotransplantation.


Subject(s)
Blood Coagulation/physiology , Galactosyltransferases/metabolism , Transplantation, Heterologous/immunology , Animals , Animals, Genetically Modified/genetics , Galactosyltransferases/genetics , Gene Knockout Techniques/methods , Genetic Engineering/methods , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Primates , Swine
18.
PLoS One ; 7(10): e47273, 2012.
Article in English | MEDLINE | ID: mdl-23118867

ABSTRACT

BACKGROUND: Baboons receiving xenogeneic livers from wild type and transgenic pigs survive less than 10 days. One of the major issues is the early development of profound thrombocytopenia that results in fatal hemorrhage. Histological examination of xenotransplanted livers has shown baboon platelet activation, phagocytosis and sequestration within the sinusoids. In order to study the mechanisms of platelet consumption in liver xenotransplantation, we have developed an in vitro system to examine the interaction between pig endothelial cells with baboon platelets and to thereby identify molecular mechanisms and therapies. METHODS: Fresh pig hepatocytes, liver sinusoidal and aortic endothelial cells were isolated by collagenase digestion of livers and processing of aortae from GTKO and Gal+ MGH-miniature swine. These primary cell cultures were then tested for the differential ability to induce baboon or pig platelet aggregation. Phagocytosis was evaluated by direct observation of CFSE labeled-platelets, which are incubated with endothelial cells under confocal light microscopy. Aurintricarboxylic acid (GpIb antagonist blocking interactions with von Willebrand factor/vWF), eptifibatide (Gp IIb/IIIa antagonist), and anti-Mac-1 Ab (anti-α(M)ß(2) integrin Ab) were tested for the ability to inhibit phagocytosis. RESULTS: None of the pig cells induced aggregation or phagocytosis of porcine platelets. However, pig hepatocytes, liver sinusoidal and aortic endothelial cells (GTKO and Gal+) all induced moderate aggregation of baboon platelets. Importantly, pig liver sinusoidal endothelial cells efficiently phagocytosed baboon platelets, while pig aortic endothelial cells and hepatocytes had minimal effects on platelet numbers. Anti-MAC-1 Ab, aurintricarboxylic acid or eptifibatide, significantly decreased baboon platelet phagocytosis by pig liver endothelial cells (P<0.01). CONCLUSIONS: Although pig hepatocytes and aortic endothelial cells directly caused aggregation of baboon platelets, only pig liver endothelial cells efficiently phagocytosed baboon platelets. Blocking vWF and integrin adhesion pathways prevented both aggregation and phagocytosis.


Subject(s)
Endothelial Cells , Platelet Aggregation , Swine , Transplantation, Heterologous , von Willebrand Factor , Animals , Aurintricarboxylic Acid/administration & dosage , Endothelial Cells/cytology , Endothelial Cells/metabolism , Eptifibatide , Hepatocytes/immunology , Hepatocytes/metabolism , Liver Transplantation/adverse effects , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/metabolism , Papio/immunology , Papio/physiology , Peptides/administration & dosage , Phagocytosis/genetics , Phagocytosis/immunology , Platelet Aggregation/genetics , Platelet Aggregation/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Swine/genetics , Swine/immunology , Transplantation, Heterologous/adverse effects , Transplantation, Heterologous/immunology , von Willebrand Factor/antagonists & inhibitors , von Willebrand Factor/metabolism
20.
Expert Rev Clin Immunol ; 8(7): 621-34, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23078060

ABSTRACT

Pigs are currently the preferred species for future organ xenotransplantation. With advances in the development of genetically modified pigs, clinical xenotransplantation is becoming closer to reality. In preclinical studies (pig-to-nonhuman primate), the xenotransplantation of livers from pigs transgenic for human CD55 or from α1,3-galactosyltransferase gene-knockout pigs+/- transgenic for human CD46, is associated with survival of approximately 7-9 days. Although hepatic function, including coagulation, has proved to be satisfactory, the immediate development of thrombocytopenia is very limiting for pig liver xenotransplantation even as a 'bridge' to allotransplantation. Current studies are directed to understand the immunobiology of platelet activation, aggregation and phagocytosis, in particular the interaction between platelets and liver sinusoidal endothelial cells, hepatocytes and Kupffer cells, toward identifying interventions that may enable clinical application.


Subject(s)
Liver Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Animals, Genetically Modified , Blood Platelets/immunology , Humans , Liver/cytology , Liver/metabolism , Liver Transplantation/adverse effects , Swine , Transplantation, Heterologous/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...