Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 17218, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37821577

ABSTRACT

Transgenic expression of protective molecules in porcine cells and tissues is a promising approach to prevent xenograft rejection. Viruses have developed various strategies to escape the host's immune system. We generated porcine B cells (B cell line L23) expressing the human adenovirus protein E3/49K or the human cytomegalovirus protein pUL11 and investigated how human T, NK and B cell responses are affected by the expression of the viral proteins. Binding studies revealed that E3/49K and pUL11 interact with CD45 on human but not porcine peripheral blood mononuclear cells. T cell proliferation in response to L23-E3/49K cells was significantly reduced and accompanied by development of an anti-inflammatory cytokine milieu (low: TNF-alpha, IFN-gamma, IL-6; high: IL-4, IL-10). Human peripheral blood mononuclear cells which had been primed for four weeks by L23-E3/49K cells included an extended population of regulatory T cells. Cytotoxicity of effector T and natural killer cells against L23 cells was significantly reduced (40 to 50%) by E3/49K expression. B cell activation and antibody production to E3/49K expressing cells was also diminished. Surprisingly, pUL11 expression showed no effects. Reduction of human anti-pig immune responses by transgenic expression of selected viral genes may be a novel approach for protection of porcine xenografts.


Subject(s)
Killer Cells, Natural , Leukocytes, Mononuclear , Animals , Humans , Swine , Leukocytes, Mononuclear/metabolism , Ligands , Killer Cells, Natural/metabolism , Cells, Cultured , Animals, Genetically Modified , Cytomegalovirus/metabolism , Viral Proteins/genetics , Immunity
2.
Am J Transplant ; 20(4): 988-998, 2020 04.
Article in English | MEDLINE | ID: mdl-31733031

ABSTRACT

Porcine xenografts lacking swine leukocyte antigen (SLA) class I are thought to be protected from human T cell responses. We have previously shown that SLA class I deficiency can be achieved in pigs by CRISPR/Cas9-mediated deletion of ß2 -microglobulin (B2M). Here, we characterized another line of genetically modified pigs in which targeting of the B2M locus did not result in complete absence of B2M and SLA class I but rather in significantly reduced expression levels of both molecules. Residual SLA class I was functionally inert, because no proper differentiation of the CD8+ T cell subset was observed in B2Mlow pigs. Cells from B2Mlow pigs were less capable in triggering proliferation of human peripheral blood mononuclear cells in vitro, which was mainly due to the nonresponsiveness of CD8+ T cells. Nevertheless, cytotoxic effector cells developing from unaffected cell populations (eg, CD4+ T cells, natural killer cells) lysed targets from both SLA class I+ wildtype and SLA class Ilow pigs with similar efficiency. These data indicate that the absence of SLA class I is an effective approach to prevent the activation of human CD8+ T cells during the induction phase of an anti-xenograft response. However, cytotoxic activity of cells during the effector phase cannot be controlled by this approach.


Subject(s)
CD8-Positive T-Lymphocytes , Leukocytes, Mononuclear , Animals , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II , Humans , Immunity , Phenotype , Swine
3.
Xenotransplantation ; 26(6): e12525, 2019 11.
Article in English | MEDLINE | ID: mdl-31119817

ABSTRACT

BACKGROUND: Despite major improvements in pig-to-primate xenotransplantation, long-term survival of xenografts is still challenging. The major histocompatibility complex (MHC) class I, which is crucial in cellular immune response, is an important xenoantigen. Abrogating MHC class I expression on xenografts might be beneficial for extending graft survival beyond current limits. METHODS: In this study, we employed the CRISPR/Cas9 system to target exon 2 of the porcine beta-2-microglobulin (B2M) gene to abrogate SLA class I expression on porcine cells. B2M-KO cells served as donor cells for somatic cell nuclear transfer, and cloned embryos were transferred to three recipient sows. The offspring were genotyped for mutations at the B2M locus, and blood samples were analyzed via flow cytometry for the absence of SLA class I molecules. RESULTS: Pregnancies were successfully established and led to the birth of seven viable piglets. Genomic sequencing proved that all piglets carried biallelic modifications at the B2M locus leading to a frameshift, a premature stop codon, and ultimately a functional knockout. However, survival times of these animals did not exceed 4 weeks due to unexpected disease processes. CONCLUSION: Here, we demonstrate the feasibility of generating SLA class I knockout pigs by targeting the porcine beta-2-microglobulin gene using the CRISPR/Cas9 system. Additionally, our findings indicate for the first time that this genetic modification might have a negative impact on the viability of the animals. These issues need to be solved to unveil the real value for xenotransplantation in the future.


Subject(s)
Galactosyltransferases/genetics , Histocompatibility Antigens Class I/genetics , Transplantation, Heterologous , beta 2-Microglobulin/genetics , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Female , Gene Knockout Techniques/methods , Nuclear Transfer Techniques , Pregnancy , Swine , Transplantation, Heterologous/methods
4.
Xenotransplantation ; 25(5): e12387, 2018 09.
Article in English | MEDLINE | ID: mdl-29446180

ABSTRACT

BACKGROUND: The programmed cell death-1 (PD-1, CD279)/PD-Ligand1 (PD-L1, CD274) receptor system is crucial for controlling the balance between immune activation and induction of tolerance via generation of inhibitory signals. Expression of PD-L1 is associated with reduced immunogenicity and renders cells and tissues to an immune-privileged/tolerogenic state. METHODS: To apply this concept for clinical xenotransplantation, we generated human (h)PD-L1 transgenic pigs and characterized expression and biological function of the transgene at the cellular level. RESULTS: The hPD-L1 was detected in kidney, heart, and pancreas. In addition, peripheral blood mononuclear cells (PBMC), cultured fibroblasts, and endothelial cells were hPD-L1 positive (hPD-L1+ ). The hPD-L1 levels were increased by the treatment of transgenic cells with human cytokines (eg, TNF-α), suggesting a regulatable mode of transgene expression. Compared to cells from wild-type pigs, hPD-L1+ PBMC had a significantly reduced capacity to stimulate proliferation of human CD4+ T cells. Moreover, fibroblasts from hPD-L1 transgenic pigs were partially protected from cell-mediated lysis by human cytotoxic effector cells. CONCLUSIONS: These data indicate a low immunogenic, immune-protected status of cells from hPD-L1 transgenic pigs. The integration of the hPD-L1 concept into existing multi-transgenic pigs is promising to achieve long-term survival of porcine xenografts in non-human primate recipients.


Subject(s)
Animals, Genetically Modified/immunology , B7-H1 Antigen/metabolism , Heterografts/immunology , Leukocytes, Mononuclear/immunology , T-Lymphocytes/immunology , Animals , Antibody Formation/immunology , Cell Proliferation/physiology , Cytotoxicity, Immunologic/immunology , Endothelial Cells/immunology , Humans , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Swine , Transplantation, Heterologous
5.
Xenotransplantation ; 25(2): e12382, 2018 03.
Article in English | MEDLINE | ID: mdl-29359453

ABSTRACT

BACKGROUND: Multiple xenoprotective transgenes are best grouped at a single locus to avoid segregation during breeding and simplify production of donor animals. METHODS: We used transgene stacking to place a human CD55 transgene adjacent to a human heme oxygenase 1 construct at the porcine ROSA26 locus. A transgenic pig was analyzed by PCR, RT-PCR, droplet digital PCR, immunohistochemistry, immunofluorescence, and flow cytometry. Resistance to complement-mediated cell lysis and caspase 3/7 activation were determined in vitro. RESULTS: The ROSA26 locus was retargeted efficiently, and animals were generated by nuclear transfer. RNA and protein analyses revealed abundant expression in all organs analyzed, including pancreatic beta cells. Transgenic porcine kidney fibroblasts were almost completely protected against complement-mediated lysis and showed reduced caspase 3/7 activation. CONCLUSION: Step-by-step placement enables highly expressed single-copy xenoprotective transgenes to be grouped at porcine ROSA26.


Subject(s)
Insulin-Secreting Cells/cytology , Transplantation, Heterologous , Animals , Animals, Genetically Modified/genetics , CD55 Antigens/genetics , CD59 Antigens/genetics , Fibroblasts/cytology , Genetic Loci , Heme Oxygenase-1/genetics , Humans , Promoter Regions, Genetic/genetics , Swine , Transgenes/genetics , Transplantation, Heterologous/methods
6.
Xenotransplantation ; 23(5): 347-56, 2016 09.
Article in English | MEDLINE | ID: mdl-27613101

ABSTRACT

BACKGROUND: The development of donor-reactive antibodies is regarded to be an important barrier limiting long-term outcome of allo- and xenografts. We asked whether enhanced signaling via the co-inhibitory receptor programmed cell death-1 (PD-1; CD279) can downregulate human B-cell activation. METHODS: Proliferation of human purified CD19(+) B cells was induced by in vitro stimulation with CpG oligodeoxynucleotides (CpG-B). To induce antibody production, peripheral blood mononuclear cells were co-cultured with the porcine B-cell line L23. Triggering of inhibitory signals via the PD-1 receptor was obtained either using a recombinant agonistic soluble ligand (PD-L1.Ig) or L23 transfectants overexpressing membrane-bound human PD-L1 (CD274; L23-PD-L1 cells). RESULTS: Stimulation of purified CD19(+) B cells with CpG-B resulted in upregulation of PD-1 and strong proliferation. Addition of PD-L1.Ig significantly reduced B-cell proliferation in a dose-dependent manner. A great proportion (~1%) of human circulating B cells recognizes the epitope galactose-α1,3-galactose-ß1,4-N-acetylglucosamine-R (α-gal). Thus, when B cells-in the presence of T cell help-were cocultured with α-gal-expressing L23 cells, anti-gal and anti-L23 antibodies could readily be detected in the culture supernatant. The level of induced antibodies was significantly reduced when stimulation was performed by L23-PD-L1 cells. CONCLUSIONS: Enhancing inhibitory signals may be part of future protocols to better control humoral immunity to allo- and xenografts.


Subject(s)
B-Lymphocytes/immunology , B7-H1 Antigen/immunology , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , Animals , Cell Proliferation/physiology , Cells, Cultured , Coculture Techniques , Humans , Swine , T-Lymphocytes/immunology
7.
Sci Rep ; 6: 29081, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27353424

ABSTRACT

Xenotransplantation from pigs could alleviate the shortage of human tissues and organs for transplantation. Means have been identified to overcome hyperacute rejection and acute vascular rejection mechanisms mounted by the recipient. The challenge is to combine multiple genetic modifications to enable normal animal breeding and meet the demand for transplants. We used two methods to colocate xenoprotective transgenes at one locus, sequential targeted transgene placement - 'gene stacking', and cointegration of multiple engineered large vectors - 'combineering', to generate pigs carrying modifications considered necessary to inhibit short to mid-term xenograft rejection. Pigs were generated by serial nuclear transfer and analysed at intermediate stages. Human complement inhibitors CD46, CD55 and CD59 were abundantly expressed in all tissues examined, human HO1 and human A20 were widely expressed. ZFN or CRISPR/Cas9 mediated homozygous GGTA1 and CMAH knockout abolished α-Gal and Neu5Gc epitopes. Cells from multi-transgenic piglets showed complete protection against human complement-mediated lysis, even before GGTA1 knockout. Blockade of endothelial activation reduced TNFα-induced E-selectin expression, IFNγ-induced MHC class-II upregulation and TNFα/cycloheximide caspase induction. Microbial analysis found no PERV-C, PCMV or 13 other infectious agents. These animals are a major advance towards clinical porcine xenotransplantation and demonstrate that livestock engineering has come of age.


Subject(s)
Complement System Proteins/genetics , Gene Editing , Heterografts , Transplantation, Heterologous/methods , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/immunology , CRISPR-Cas Systems/genetics , Complement System Proteins/immunology , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Swine/genetics , Swine/immunology
8.
PLoS One ; 11(5): e0155676, 2016.
Article in English | MEDLINE | ID: mdl-27175998

ABSTRACT

We have successfully established and characterized a genetically modified pig line with ubiquitous expression of LEA29Y, a human CTLA4-Ig derivate. LEA29Y binds human B7.1/CD80 and B7.2/CD86 with high affinity and is thus a potent inhibitor of T cell co-stimulation via this pathway. We have characterized the expression pattern and the biological function of the transgene as well as its impact on the porcine immune system and have evaluated the potential of these transgenic pigs to propagate via assisted breeding methods. The analysis of LEA29Y expression in serum and multiple organs of CAG-LEA transgenic pigs revealed that these animals produce a biologically active transgenic product at a considerable level. They present with an immune system affected by transgene expression, but can be maintained until sexual maturity and propagated by assisted reproduction techniques. Based on previous experience with pancreatic islets expressing LEA29Y, tissues from CAG-LEA29Y transgenic pigs should be protected against rejection by human T cells. Furthermore, their immune-compromised phenotype makes CAG-LEA29Y transgenic pigs an interesting large animal model for testing human cell therapies and will provide an important tool for further clarifying the LEA29Y mode of action.


Subject(s)
Abatacept/metabolism , Lymphocyte Activation/immunology , Reproduction/genetics , Sus scrofa/genetics , Sus scrofa/immunology , T-Lymphocytes/immunology , Animals , Animals, Genetically Modified , Antigen-Presenting Cells/metabolism , Cloning, Organism , Conserved Sequence , Crosses, Genetic , Female , Fertilization in Vitro , Humans , Lymph Nodes/pathology , Male , Promoter Regions, Genetic/genetics , Protein Binding
9.
Transplant Direct ; 1(6): e23, 2015 Jul.
Article in English | MEDLINE | ID: mdl-27500225

ABSTRACT

UNLABELLED: Multiple modifications of the porcine genome are required to prevent rejection after pig-to-primate xenotransplantation. Here, we produced pigs with a knockout of the α1,3-galactosyltransferase gene (GGTA1-KO) combined with transgenic expression of the human anti-apoptotic/anti-inflammatory molecules heme oxygenase-1 and A20, and investigated their xenoprotective properties. METHODS: The GGTA1-KO/human heme oxygenase-1 (hHO-1)/human A20 (hA20) transgenic pigs were produced in a stepwise approach using zinc finger nuclease vectors targeting the GGTA1 gene and a Sleeping Beauty vector coding for hA20. Two piglets were analyzed by quantitative reverse-transcription polymerase chain reaction, flow cytometry, and sequencing. The biological function of the genetic modifications was tested in a (51)Chromium release assay and by ex vivo kidney perfusions with human blood. RESULTS: Disruption of the GGTA1 gene by deletion of few basepairs was demonstrated in GGTA1-KO/hHO-1/hA20 transgenic pigs. The hHO-1 and hA20 mRNA expression was confirmed by quantitative reverse-transcription polymerase chain reaction. Ex vivo perfusion of 2 transgenic kidneys was feasible for the maximum experimental time of 240 minutes without symptoms of rejection. CONCLUSIONS: Results indicate that GGTA1-KO/hHO-1/hA20 transgenic pigs are a promising model to alleviate rejection and ischemia-reperfusion damage in porcine xenografts and could serve as a background for further genetic modifications toward the production of a donor pig that is clinically relevant for xenotransplantation.

10.
Transplantation ; 97(2): 138-47, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24150517

ABSTRACT

BACKGROUND: Among other mismatches between human and pig, incompatibilities in the blood coagulation systems hamper the xenotransplantation of vascularized organs. The provision of the porcine endothelium with human thrombomodulin (hTM) is hypothesized to overcome the impaired activation of protein C by a heterodimer consisting of human thrombin and porcine TM. METHODS: We evaluated regulatory regions of the THBD gene, optimized vectors for transgene expression, and generated hTM expressing pigs by somatic cell nuclear transfer. Genetically modified pigs were characterized at the molecular, cellular, histological, and physiological levels. RESULTS: A 7.6-kb fragment containing the entire upstream region of the porcine THBD gene was found to drive a high expression in a porcine endothelial cell line and was therefore used to control hTM expression in transgenic pigs. The abundance of hTM was restricted to the endothelium, according to the predicted pattern, and the transgene expression of hTM was stably inherited to the offspring. When endothelial cells from pigs carrying the hTM transgene--either alone or in combination with an aGalTKO and a transgene encoding the human CD46-were tested in a coagulation assay with human whole blood, the clotting time was increased three- to four-fold (P<0.001) compared to wild-type and aGalTKO/CD46 transgenic endothelial cells. This, for the first time, demonstrated the anticoagulant properties of hTM on porcine endothelial cells in a human whole blood assay. CONCLUSIONS: The biological efficacy of hTM suggests that the (multi-)transgenic donor pigs described here have the potential to overcome coagulation incompatibilities in pig-to-primate xenotransplantation.


Subject(s)
Animals, Genetically Modified , Endothelial Cells/metabolism , Regulatory Sequences, Nucleic Acid , Swine/genetics , Thrombomodulin/genetics , Animals , Genetic Vectors , Humans , Membrane Cofactor Protein/analysis , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Thrombomodulin/physiology , Transplantation, Heterologous
11.
Xenotransplantation ; 19(1): 40-51, 2012.
Article in English | MEDLINE | ID: mdl-22360752

ABSTRACT

BACKGROUND: Efficient and precise techniques for the genetic modification of pigs facilitate the generation of tailored donor animals for xenotransplantation. Numerous transgenic pig lines exist with the focus on inhibition of the complement system and of humoral immune responses. In addition, immune cell-based responses need to be controlled to prevent pig-to-primate xenograft rejection. Expression of human (hu) TNF-related apoptosis-inducing ligand (TRAIL) on porcine cells has the potential to ameliorate human T cell responses. METHODS: We generated transgenic pigs expressing human tumor necrosis factor (TNF)-related apoptosis-inducing ligand (huTRAIL) under the control of either the mouse H2K(b) promoter or a CMV enhancer/chicken ß-actin (CAG) promoter, the latter one (CAG-huTRAIL) on a GGTA1 knockout/huCD46 transgenic background. The biological activity of huTRAIL was demonstrated by its apoptosis-inducing effect on Jurkat lymphoma cells. To clarify whether huTRAIL affects also primary immune cells and whether its effects depend on the presence of co-stimulatory molecules, we exposed human peripheral blood mononuclear cells (PBMC) or isolated T cells to huTRAIL-expressing porcine fibroblasts or dendritic cells in vitro. RESULTS: H2Kb-huTRAIL transgenic pigs express huTRAIL mainly in the spleen and secondary lymphoid tissues. The CAG-huTRAIL construct facilitated huTRAIL expression in multiple organs, the level being at least one order of magnitude higher than in H2Kb-huTRAIL transgenic pigs. Incubation with huTRAIL-expressing H2Kb-huTRAIL transgenic porcine dendritic cells decreased human T cell proliferation significantly without any signs of apoptosis. In spite of the high transgene expression level, CAG-huTRAIL transgenic fibroblasts did not affect proliferation of human PBMC, independent of their activation state. CONCLUSIONS: These results suggest huTRAIL expression on porcine dendritic cells as a possible strategy to attenuate T cell responses against pig-to-primate xenografts.


Subject(s)
Dendritic Cells/metabolism , T-Lymphocytes/immunology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transplantation, Heterologous/immunology , Animals , Animals, Genetically Modified , Apoptosis/physiology , Cell Proliferation , Cells, Cultured , Humans , Jurkat Cells , Leukocytes, Mononuclear/immunology , Swine
12.
Xenotransplantation ; 18(6): 355-68, 2011.
Article in English | MEDLINE | ID: mdl-22168142

ABSTRACT

BACKGROUND: The major immunological hurdle to successful porcine-to-human xenotransplantation is the acute vascular rejection (AVR), characterized by endothelial cell (EC) activation and perturbation of coagulation. Heme oxygenase-1 (HO-1) and its derivatives have anti-apoptotic, anti-inflammatory effects and protect against reactive oxygen species, rendering HO-1 a promising molecule to control AVR. Here, we report the production and characterization of pigs transgenic for human heme oxygenase-1 (hHO-1) and demonstrate significant protection in porcine kidneys against xenograft rejection in ex vivo perfusion with human blood and transgenic porcine aortic endothelial cells (PAEC) in a TNF-α-mediated apoptosis assay. METHODS: Transgenic and non-transgenic PAEC were tested in a TNF-α-mediated apoptosis assay. Expression of adhesion molecules (ICAM-1, VCAM-1, and E-selectin) was measured by real-time PCR. hHO-1 transgenic porcine kidneys were perfused with pooled and diluted human AB blood in an ex vivo perfusion circuit. MHC class-II up-regulation after induction with IFN-γ was compared between wild-type and hHO-1 transgenic PAEC. RESULTS: Cloned hHO-1 transgenic pigs expressed hHO-1 in heart, kidney, liver, and in cultured ECs and fibroblasts. hHO-1 transgenic PAEC were protected against TNF-α-mediated apoptosis. Real-time PCR revealed reduced expression of adhesion molecules like ICAM-1, VCAM-1, and E-selectin. These effects could be abrogated by the incubation of transgenic PAECs with the specific HO-1 inhibitor zinc protoporphorine IX (Zn(II)PPIX, 20 µm). IFN-γ induced up-regulation of MHC class-II molecules was significantly reduced in PAECs from hHO-1 transgenic pigs. hHO-1 transgenic porcine kidneys could successfully be perfused with diluted human AB-pooled blood for a maximum of 240 min (with and without C1 inh), while in wild-type kidneys, blood flow ceased after ∼60 min. Elevated levels of d-Dimer and TAT were detected, but no significant consumption of fibrinogen and antithrombin was determined. Microthrombi could not be detected histologically. CONCLUSIONS: These results are encouraging and warrant further studies on the biological function of heme oxygenase-I expression in hHO-1 transgenic pigs in the context of xenotransplantation.


Subject(s)
Graft Rejection/prevention & control , Heme Oxygenase-1/metabolism , Kidney/immunology , Transplantation, Heterologous/immunology , Animals , Animals, Genetically Modified , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/physiology , Graft Rejection/immunology , Heme Oxygenase-1/genetics , Humans , Kidney/blood supply , Kidney/physiology , Perfusion , Swine , Transgenes
13.
Transpl Int ; 23(12): 1293-300, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20579316

ABSTRACT

Cellular rejection is a relevant hurdle for successful pig-to-primate xenotransplantation. We have shown previously that the induction of a human anti-pig T cell response (in vitro activation of CD4(+) T cells) can be suppressed by the overexpression of human negative costimulatory ligands (e.g. programmed death receptor ligand, PD-L1) on pig antigen presenting cells. Here, we asked whether PD-L1 mediated enhancement of negative signaling might also be efficient during the effector phase of human anti-pig cellular immune responses. The porcine B-cell line L23 was transfected with human PD-L1, and clones were selected stably expressing PD-L1 with low, medium, or high density. Mock-transfected L23 cells were effectively lysed by human cytotoxic effector cells (IL-2 activated CD8(+) T cells and CD56(+) cells). The lytic potential of the effectors decreased with increasing levels of PD-L1 and was reduced by about 50% in L23-PD-L(high) targets. A proportion of activated CD8(+) effector cells underwent apoptosis when exposed to PD-L1 expressing L23 cells. These data suggest that the overexpression of PD-L1 on target cells may (a) trigger negative signals in effector cells that prevent the release of cytolytic molecules and/or (b) induce apoptosis in the attacking effector cells thereby protecting targets from destruction.


Subject(s)
Antigens, CD/biosynthesis , Graft Rejection/immunology , Sus scrofa/immunology , Transplantation, Heterologous/immunology , Animals , B-Lymphocytes/immunology , B7-H1 Antigen , CD56 Antigen/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/immunology , Down-Regulation , Humans , Transfection
14.
Transplantation ; 87(7): 975-82, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19352115

ABSTRACT

BACKGROUND: Genetic modification of pigs (e.g., transgenic expression of human complement regulatory molecules or inactivation of alpha1,3galactosyltransferase) enabled the development of promising strategies to overcome hyperacute rejection after pig-to-primate xenotransplantation. However, cellular rejection still remains a hurdle for successful xenograft survival. This report tested the hypothesis that overexpression of human negative costimulatory PD-Ligands (PD-L) in pig antigen presenting cells might be an approach to prevent human anti-pig T-cell responses. METHODS: The pig B-cell line L23 was transfected with the pIRES-AcGFP vector containing human PD-L1 or PD-L2. Stable transfectants (L23-PD-L1, L23-PD-L2 cells) were established and used for in vitro stimulation of purified human CD4+ T cells. RESULTS: Human CD4+ T cells responded with significantly reduced proliferation to L23-PD-L1 or L23-PD-L2 cells and produced less IL-2, IFNgamma, TNFalpha, IL-4, and IL-5 than cells stimulated with mock-transfected B cells. The concentration of IL-10, however, was increased in CD4+ T cells responding to stimulation with PD-L1 or PD-L2 transfectants. Furthermore, in cultures of CD4+ T cells stimulated for 3 weeks with PD-L1 or PD-L2 transfectants a CD4+CD25(high)Foxp3+ subset showed up that effectively suppressed the activation of conventional CD4+ T cells. CONCLUSIONS: These findings imply that PD-1/PD-Ligand pathways are interesting targets to prevent human anti-pig T-cell responses after xenotransplantation, and also suggests that PD-1/PD-Ligand interactions may play a role in the control of the activity and/or homeostasis of regulatory T cells.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , B7-H1 Antigen , Cell Line , Flow Cytometry , Gene Amplification , Graft Survival/immunology , Humans , Immunosuppression Therapy , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Lymphocyte Activation , Polymerase Chain Reaction , Primates , Programmed Cell Death 1 Ligand 2 Protein , Swine , Transfection , Transplantation, Heterologous/immunology
15.
J Immunol ; 176(2): 931-8, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16393978

ABSTRACT

The 77C-->G mutation in exon A of the human CD45 gene occurs with low frequency in healthy individuals. An enhanced frequency of 77C-->G individuals has been reported in cohorts of patients suffering from multiple sclerosis, systemic sclerosis, autoimmune hepatitis, and HIV-1. To investigate the mechanisms by which the variant allele may contribute to disease susceptibility, we compared T cell reactivity in heterozygous carriers of the mutation (healthy individuals and multiple sclerosis patients) and wild-type controls. In vitro-generated T cell lines and freshly isolated CD4+CD45R0+ primed/memory T cells from 77C-->G individuals aberrantly expressed CD45RA isoforms and showed enhanced proliferation and IL-2 production when stimulated with anti-TCR/CD3 mAb or Ag. Mutant T cell lines contained a more active pool of p56lck tyrosine kinase and responded with increased phosphorylation of Zap70 and TCR-zeta and an enhanced Ca2+ flux to TCR/CD3 stimulation. These data suggest that 77C-->G may act as a risk factor for certain diseases by increasing the intensity of TCR signaling.


Subject(s)
Leukocyte Common Antigens/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Point Mutation , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Antibodies, Monoclonal/pharmacology , Base Sequence , Case-Control Studies , Cell Line , DNA/genetics , Heterozygote , Humans , Immunologic Memory , In Vitro Techniques , Isoantigens/administration & dosage , Signal Transduction
16.
Ann Transplant ; 8(3): 35-8, 2003.
Article in English | MEDLINE | ID: mdl-15114937

ABSTRACT

UNLABELLED: Antibody-mediated targeting of pig costimulatory molecules is assumed to be a possible strategy to achieve donor-specific tolerance after xenotransplantation. However, porcine molecules of the B7 family (e.g. CD86) are expressed on typical antigen presenting cells (APC) and also on vascular endothelial cells. Thus, in vascularized porcine xenografts the usage of therapeutic anti-B7 monoclonal antibodies (mAb) might be associated with damage of the endothelium. OBJECTIVE: In the present study we asked whether modulation of human T cell reactivity can be obtained by targeting molecules selectively expressed on pig leucocytes. METHODS: MAb directed to pig CD45 were tested for their capacity to modulate the in vitro activation of human T cells induced by porcine peripheral blood mononuclear cells. RESULTS: Porcine stimulatory cells induced significant proliferation of human T cells. In the presence of porcine CD45 mAb human T cell responses were reduced by 30-40%. The inhibitory effects were most pronounced when CD45RA mAb were used whereas mAb directed to CD45RC isoforms only moderately inhibited human T cell activation. The tested antibodies had no effects on human T cell activation induced by mitogens or by alloantigen. CONCLUSION: Manipulation of CD45 molecules on pig leucocytes may reduce their potential to stimulate human T cells. In recipients of vascularized porcine xenografts the usage of anti-pig CD45 mAb could be an approach to block the direct pathway of T cell activation initiated by porcine APC without affecting the endothelium of the graft.


Subject(s)
Antibodies, Heterophile/immunology , Leukocyte Common Antigens/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , Antigen-Presenting Cells/immunology , Endothelium, Vascular/transplantation , Humans , Immunosuppression Therapy , Lymphocyte Activation/immunology , Swine , Transplantation, Heterologous/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...