Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Front Immunol ; 15: 1279050, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38352884

RESUMEN

Xenotransplantation offers a promising alternative to circumvent the lack of donated human organs available for transplantation. Different attempts to improve the survival of xenografts led to the generation of transgenic pigs expressing various combinations of human protective genes or knocked out for specific antigens. Currently, testing the efficiency of porcine organs carrying different genetic modifications in preventing xenogeneic immune responses completely relies on in vitro assays, humanized mouse models, or non-human primate transplantation models. However, these tests are often associated with major concerns due to reproducibility and generation of insufficient data as well as they raise ethical, logistical, and economic issues. In this study, we investigated the feasibility of specifically assessing the strength of human T-cell responses towards the kidneys of wild-type (WT) or transgenic pigs overexpressing human programmed death-1 ligand 1 (hPD-L1) during ex vivo kidney perfusion (EVKP). Human T cells were shown to adhere to the endothelium and transmigrate into WT and hPD-L1 kidneys. However, transcript levels of TNF-a and IFN-y as well as cytotoxic molecules such as granzyme B and perforin secreted by human T cells were significantly decreased in the tissue of hPD-L1 kidneys in comparison to WT kidneys. These results were confirmed via in vitro assays using renal endothelial cells (ECs) isolated from WT and hPD-L1 transgenic pigs. Both CD4+ and CD8+ T cells showed significantly lower proliferation rates after exposure to hPD-L1 porcine renal ECs in comparison to WT ECs. In addition, the secretion of pro-inflammatory cytokines was significantly reduced in cultures using hPD-L1 ECs in comparison to WT ECs. Remarkably, hPD-L1 EC survival was significantly increased in cytotoxic assays. This study demonstrates the feasibility of evaluating the human response of specific immune subsets such as human T cells towards the whole xenograft during EVKP. This may represent a robust strategy to assess the potency of different genetic modifications to prevent xenogeneic immune responses and thereby predict the risk of immune rejection of new genetically engineered xenografts.


Asunto(s)
Antígeno B7-H1 , Linfocitos T CD8-positivos , Ratones , Animales , Porcinos , Humanos , Antígeno B7-H1/genética , Células Endoteliales , Reproducibilidad de los Resultados , Animales Modificados Genéticamente , Activación de Linfocitos , Riñón
2.
Am J Transplant ; 24(1): 20-29, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37659605

RESUMEN

Strong xenorejection limits the clinical application of porcine islet transplantation in type 1 diabetes. Targeting T cell-mediated rejection is one of the main approaches to improve long-term graft survival. Here we study engraftment and survival of porcine islet cells expressing human programmed cell death ligand-1 (hPD-L1) in a humanized mouse model. Neonatal islet-like clusters (NPICCs) from transgenic hPD-L1 (hPD-L1-Tg) and wild-type (Wt) pigs were transplanted into nonobese diabetic-scid IL2rγnull mice stably reconstituted with human immune cells (hPD-L1 n = 10; Wt n = 6). Primary endpoint was development of normoglycemia during a 16-week observation period after transplantation. Secondary endpoints were porcine C-peptide levels and immune cell infiltration. Animals transplanted with hPD-L1-Tg neonatal islet-like clusters achieved a superior normoglycemic rate (50% versus 0%) and significantly higher plasma C-peptide levels as compared to the Wt group, indicating long-term beta cell function. Intracytoplasmic fluorescence-activated cell sorting analysis and immunohistochemistry revealed significantly decreased frequencies of interferonγ-expressing splenic hCD8-positive T cells and reduced intragraft-infiltrating immune cells. We here demonstrate that expression of hPD-L1 provides strong islet xenograft protection without administration of immunosuppressive drugs. These findings support the hypothesis that hPD-L1 has the capacity to control cellular rejection and therefore represents a very promising transgene candidate for clinical porcine islet xenotransplantation.


Asunto(s)
Diabetes Mellitus , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Ratones , Animales , Humanos , Porcinos , Antígeno B7-H1/metabolismo , Péptido C/metabolismo , Islotes Pancreáticos/metabolismo , Ratones Noqueados , Trasplante Heterólogo , Ratones SCID , Rechazo de Injerto/etiología
4.
Artículo en Inglés | MEDLINE | ID: mdl-38154473

RESUMEN

This report comprises the contents of the presentations and following discussions of a workshop of the German Heart Transplant Centers in Martinsried, Germany on cardiac xenotransplantation. The production and current availability of genetically modified donor pigs, preservation techniques during organ harvesting, and immunosuppressive regimens in the recipient are described. Selection criteria for suitable patients and possible solutions to the problem of overgrowth of the xenotransplant are discussed. Obviously microbiological safety for the recipient and close contacts is essential, and ethical considerations to gain public acceptance for clinical applications are addressed. The first clinical trial will be regulated and supervised by the Paul-Ehrlich-Institute as the National Competent Authority for Germany, and the German Heart Transplant Centers agreed to cooperatively select the first patients for cardiac xenotransplantation.

5.
Sci Rep ; 13(1): 17218, 2023 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-37821577

RESUMEN

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.


Asunto(s)
Células Asesinas Naturales , Leucocitos Mononucleares , Animales , Humanos , Porcinos , Leucocitos Mononucleares/metabolismo , Ligandos , Células Asesinas Naturales/metabolismo , Células Cultivadas , Animales Modificados Genéticamente , Citomegalovirus/metabolismo , Proteínas Virales/genética , Inmunidad
6.
Int J Mol Sci ; 24(16)2023 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-37628892

RESUMEN

Xenotransplantation reemerged as a promising alternative to conventional transplantation enlarging the available organ pool. However, success of xenotransplantation depends on the design and selection of specific genetic modifications and on the development of robust assays allowing for a precise assessment of tissue-specific immune responses. Nevertheless, cell-based assays are often compromised by low proliferative capacity of primary cells. Proximal tubular epithelial cells (PTECs) play a crucial role in kidney function. Here, we generated immortalized PTECs (imPTECs) by overexpression of simian virus 40 T large antigen. ImPTECs not only showed typical morphology and phenotype, but, in contrast to primary PTECs, they maintained steady cell cycling rates and functionality. Furthermore, swine leukocyte antigen (SLA) class I and class II transcript levels were reduced by up to 85% after transduction with lentiviral vectors encoding for short hairpin RNAs targeting ß2-microglobulin and the class II transactivator. This contributed to reducing xenogeneic T-cell cytotoxicity (p < 0.01) and decreasing secretion of pro-inflammatory cytokines such as IL-6 and IFN-γ. This study showed the feasibility of generating highly proliferative PTECs and the development of tissue-specific immunomonitoring assays. Silencing SLA expression on PTECs was demonstrated to be an effective strategy to prevent xenogeneic cellular immune responses and may strongly support graft survival after xenotransplantation.


Asunto(s)
Bioensayo , Células Epiteliales , Animales , Porcinos , Regulación hacia Abajo , Inmunidad
7.
Transplantation ; 105(9): 1930-1943, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33350675

RESUMEN

Heart transplantation is the only long-lasting lifesaving option for patients with terminal cardiac failure. The number of available human organs is however far below the actual need, resulting in substantial mortality of patients while waiting for a human heart. Mechanical assist devices are used to support cardiac function but are associated with a high risk of severe complications and poor quality of life for the patients. Consistent success in orthotopic transplantation of genetically modified pig hearts into baboons indicates that cardiac xenotransplantation may become a clinically applicable option for heart failure patients who cannot get a human heart transplant. In this overview, we project potential paths to clinical cardiac xenotransplantation, including the choice of genetically modified source pigs; associated requirements of microbiological, including virological, safety; optimized matching of source pig and recipient; and specific treatments of the donor heart after explantation and of the recipients. Moreover, selection of patients and the regulatory framework will be discussed.


Asunto(s)
Insuficiencia Cardíaca/cirugía , Trasplante de Corazón , Donantes de Tejidos/provisión & distribución , Animales , Animales Modificados Genéticamente , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/fisiopatología , Trasplante de Corazón/efectos adversos , Humanos , Complicaciones Posoperatorias/etiología , Recuperación de la Función , Medición de Riesgo , Factores de Riesgo , Sus scrofa/genética , Trasplante Heterólogo , Resultado del Tratamiento , Listas de Espera
8.
J Cell Mol Med ; 24(9): 5070-5081, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32212307

RESUMEN

Xenotransplantation of pancreatic islets offers a promising alternative to overcome the shortage of allogeneic donors. Despite significant advances, either immune rejection or oxygen supply in immune protected encapsulated islets remains major bottlenecks for clinical application. To decrease xenogeneic immune responses, we generated tissue engineered swine leucocyte antigen (SLA)-silenced islet cell clusters (ICC). Single-cell suspensions from pancreatic islets were generated by enzymatic digestion of porcine ICCs. Cells were silenced for SLA class I and class II by lentiviral vectors encoding for short hairpin RNAs targeting beta2-microglobulin or class II transactivator, respectively. SLA-silenced ICCs-derived cells were then used to form new ICCs in stirred bioreactors in the presence of collagen VI. SLA class I silencing was designed to reach a level of up to 89% and class II by up to 81% on ICCs-derived cells. Xenogeneic T cell immune responses, NK cell and antibody-mediated cellular-dependent immune responses were significantly decreased in SLA-silenced cells. In stirred bioreactors, tissue engineered islets showed the typical 3D structure and insulin production. These data show the feasibility to generate low immunogenic porcine ICCs after single-cell engineering and post-transduction islet reassembling that might serve as an alternative to allogeneic pancreatic islet cell transplantation.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/metabolismo , Animales , Anticuerpos/química , Formación de Anticuerpos , Supervivencia Celular , Células Cultivadas , Silenciador del Gen , Ingeniería Genética/métodos , Inmunidad Celular , Insulina/metabolismo , Células Asesinas Naturales/metabolismo , Trasplante de Neoplasias , Páncreas/metabolismo , Interferencia de ARN , Porcinos , Linfocitos T/metabolismo , Activación Transcripcional , Trasplante Heterólogo
9.
Xenotransplantation ; 27(1): e12560, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31591751

RESUMEN

BACKGROUND: Cell surface carbohydrate antigens play a major role in the rejection of porcine xenografts. The most important for human recipients are α-1,3 Gal (Galactose-alpha-1,3-galactose) causing hyperacute rejection, also Neu5Gc (N-glycolylneuraminic acid) and Sd(a) blood group antigens both of which are likely to elicit acute vascular rejection given the known human immune status. Porcine cells with knockouts of the three genes responsible, GGTA1, CMAH and B4GALNT2, revealed minimal xenoreactive antibody binding after incubation with human serum. However, human leucocyte antigen (HLA) antibodies cross-reacted with swine leucocyte antigen class I (SLA-I). We previously demonstrated efficient generation of pigs with multiple xeno-transgenes placed at a single genomic locus. Here we wished to assess whether key xenoreactive antigen genes can be simultaneously inactivated and if combination with the multi-transgenic background further reduces antibody deposition and complement activation. METHODS: Multiplex CRISPR/Cas9 gene editing and somatic cell nuclear transfer were used to generate pigs carrying functional knockouts of GGTA1, CMAH, B4GALNT2 and SLA class I. Fibroblasts derived from one- to four-fold knockout animals, and from multi-transgenic cells (human CD46, CD55, CD59, HO1 and A20) with the four-fold knockout were used to examine the effects on human IgG and IgM binding or complement activation in vitro. RESULTS: Pigs were generated carrying four-fold knockouts of important xenoreactive genes. In vitro assays revealed that combination of all four gene knockouts reduced human IgG and IgM binding to porcine kidney cells more effectively than single or double knockouts. The multi-transgenic background combined with GGTA1 knockout alone reduced C3b/c and C4b/c complement activation to such an extent that further knockouts had no significant additional effect. CONCLUSION: We showed that pigs carrying several xenoprotective transgenes and knockouts of xenoreactive antigens can be readily generated and these modifications will have significant effects on xenograft survival.


Asunto(s)
Galactosiltransferasas/genética , Rechazo de Injerto/inmunología , Trasplante de Riñón , Oxigenasas de Función Mixta/genética , N-Acetilgalactosaminiltransferasas/genética , Animales , Anticuerpos Heterófilos/metabolismo , Sistemas CRISPR-Cas , Células Cultivadas , Proteínas del Sistema Complemento/metabolismo , Antígenos HLA/inmunología , Xenoinjertos/inmunología , Antígenos de Histocompatibilidad Clase I , Humanos , Porcinos , Trasplante Heterólogo
10.
Am J Transplant ; 20(4): 988-998, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31733031

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos , Leucocitos Mononucleares , Animales , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase II , Humanos , Inmunidad , Fenotipo , Porcinos
11.
Xenotransplantation ; 26(4): e12523, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31074044

RESUMEN

BACKGROUND: Differences in quality and strength of immune responses between individuals are mainly due to polymorphisms in major histocompatibility complex (MHC) molecules. Focusing on MHC class-II, we asked whether the intensity of human anti-pig T-cell responses is influenced by genetic variability in the human HLA-DRB1 and/or the porcine SLA-DRB1 locus. METHODS: ELISpot assays were performed using peripheral blood mononuclear cells (PBMCs) from 62 HLA-DRB1-typed blood donors as responder and the porcine B cell line L23 as stimulator cells. Based on the frequency of IFN-γ-secreting cells, groups of weak, medium, and strong responder individuals were defined. Mixed lymphocyte reaction (MLR) assays were performed to study the stimulatory capacity of porcine PBMCs expressing different SLA-DRB1 alleles. RESULTS: Concerning the MHC class-II configuration of human cells, we found a significant overrepresentation of HLA-DRB1*01 alleles in the medium/strong responder group as compared to individuals showing weak responses to stimulation with L23 cells. Evaluation of the role of MHC class-II variability in porcine stimulators revealed that cells expressing SLA-DRB1*06 alleles triggered strong proliferation in approximately 70% of humans. Comparison of amino acid sequences indicated that strong human anti-pig reactivity may be associated with a high rate of similarity between human and pig HLA/SLA-DRB1 alleles. CONCLUSION: Variability in human and porcine MHC determines the intensity of individual human anti-pig T-cell responses. MHC typing and cross-matching of prospective recipients of xenografts and donor pigs could be relevant to select for donor-recipient combinations with minimal anti-porcine immunity.


Asunto(s)
Antígenos Heterófilos/inmunología , Variación Biológica Individual , Cadenas HLA-DRB1/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Porcinos/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/inmunología , Alelos , Secuencia de Aminoácidos , Animales , Genotipo , Cadenas HLA-DRB1/genética , Haplotipos , Humanos , Interferón gamma/biosíntesis , Células Asesinas Naturales/metabolismo , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido
12.
Xenotransplantation ; 26(6): e12525, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31119817

RESUMEN

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.


Asunto(s)
Galactosiltransferasas/genética , Antígenos de Histocompatibilidad Clase I/genética , Trasplante Heterólogo , Microglobulina beta-2/genética , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas , Femenino , Técnicas de Inactivación de Genes/métodos , Técnicas de Transferencia Nuclear , Embarazo , Porcinos , Trasplante Heterólogo/métodos
13.
Sci Rep ; 9(1): 1021, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30705299

RESUMEN

Regulatory T cells (Tregs) are crucial in controlling allospecific immune responses. However, studies in human kidney recipients regarding the contribution of polyspecific Tregs have provided differing results and studies on alloreactive Tregs are missing completely. In this retrospective study, we specifically analyzed activated CD4+CD25highFOXP3+GARP+ Tregs in 17 patients of a living donor kidney transplantation cohort longitudinally over 24 months by flow cytometry (FOXP3: forkhead box protein 3, GARP: glycoprotein A repetitions predominant). We could demonstrate that Tregs of patients with end-stage renal disease (ESRD) are already pre-activated when compared to healthy controls. Furthermore, even though total CD4+CD25highFOXP3+ Treg numbers decreased in the first three months after transplantation, frequency of activated Tregs increased significantly representing up to 40% of all peripheral Tregs. In a cohort of living donor kidney transplantation recipients with stable graft function, frequencies of activated Tregs did not correlate with the occurrence of acute cellular rejection or chronic graft dysfunction. Our results will be important for clinical trials using adoptive Treg therapy after kidney transplantation. Adoptively transferred Tregs could be important to compensate the Treg loss at month 3, while they have to compete within the Treg niche with a large number of activated Tregs.


Asunto(s)
Trasplante de Riñón , Activación de Linfocitos/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Aloinjertos/inmunología , Humanos , Fallo Renal Crónico/inmunología , Recuento de Linfocitos , Diálisis Renal , Resultado del Tratamiento
14.
Xenotransplantation ; 25(5): e12387, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29446180

RESUMEN

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.


Asunto(s)
Animales Modificados Genéticamente/inmunología , Antígeno B7-H1/metabolismo , Xenoinjertos/inmunología , Leucocitos Mononucleares/inmunología , Linfocitos T/inmunología , Animales , Formación de Anticuerpos/inmunología , Proliferación Celular/fisiología , Citotoxicidad Inmunológica/inmunología , Células Endoteliales/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Activación de Linfocitos/inmunología , Porcinos , Trasplante Heterólogo
15.
Xenotransplantation ; 25(2): e12382, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29359453

RESUMEN

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.


Asunto(s)
Células Secretoras de Insulina/citología , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente/genética , Antígenos CD55/genética , Antígenos CD59/genética , Fibroblastos/citología , Sitios Genéticos , Hemo-Oxigenasa 1/genética , Humanos , Regiones Promotoras Genéticas/genética , Porcinos , Transgenes/genética , Trasplante Heterólogo/métodos
16.
Xenotransplantation ; 23(5): 347-56, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27613101

RESUMEN

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.


Asunto(s)
Linfocitos B/inmunología , Antígeno B7-H1/inmunología , Activación de Linfocitos/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal , Animales , Proliferación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Humanos , Porcinos , Linfocitos T/inmunología
17.
Sci Rep ; 6: 29081, 2016 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-27353424

RESUMEN

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.


Asunto(s)
Proteínas del Sistema Complemento/genética , Edición Génica , Xenoinjertos , Trasplante Heterólogo/métodos , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/inmunología , Sistemas CRISPR-Cas/genética , Proteínas del Sistema Complemento/inmunología , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Humanos , Porcinos/genética , Porcinos/inmunología
18.
PLoS One ; 11(5): e0155676, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27175998

RESUMEN

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.


Asunto(s)
Abatacept/metabolismo , Activación de Linfocitos/inmunología , Reproducción/genética , Sus scrofa/genética , Sus scrofa/inmunología , Linfocitos T/inmunología , Animales , Animales Modificados Genéticamente , Células Presentadoras de Antígenos/metabolismo , Clonación de Organismos , Secuencia Conservada , Cruzamientos Genéticos , Femenino , Fertilización In Vitro , Humanos , Ganglios Linfáticos/patología , Masculino , Regiones Promotoras Genéticas/genética , Unión Proteica
19.
PLoS One ; 10(8): e0135682, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26309225

RESUMEN

Studying genetic diversity of immunologically relevant molecules can improve our knowledge on their functional spectrum in normal immune responses and may also uncover a possible role of different variants in diseases. We characterized the c.503T>C polymorphism in the human KLRB1 gene (Killer cell lectin-like receptor, subfamily B, member 1) coding for the cell surface receptor CD161. CD161 is expressed by subsets of CD4+ and CD8+ T cells and the great majority of CD56+ natural killer (NK) cells, acting as inhibitory receptor in the latter population. Genotyping a cohort of 118 healthy individuals revealed 40% TT homozygotes, 46% TC heterozygotes, and 14% carriers of CC. There was no difference in the frequency of CD161 expressing CD4+ and CD8+ T cells between the different genotypes. However, the frequency of CD161+ NK cells was significantly decreased in CC carriers as compared to TT homozygotes. c.503T>C causes an amino acid exchange (p.Ile168Thr) in an extracellular loop of the CD161 receptor, which is regarded to be involved in binding of its ligand Lectin-like transcript 1 (LLT1). Binding studies using soluble LLT1-Fc on 293 transfectants over-expressing CD161 receptors from TT or CC carriers suggested diminished binding to the CC variant. Furthermore, triggering of CD161 either by LLT1 or anti-CD161 antibodies inhibited NK cell activation less effectively in cells from CC individuals than cells from TT carriers. These data suggest that the c.503T>C polymorphism is associated with structural alterations of the CD161 receptor. The regulation of NK cell homeostasis and activation apparently differs between carriers of the CC and TT variant of CD161.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/genética , Subfamilia B de Receptores Similares a Lectina de Células NK/genética , Secuencia de Bases , Células Cultivadas , Clonación Molecular , Frecuencia de los Genes , Genotipo , Células HEK293 , Humanos , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Polimorfismo de Nucleótido Simple , Unión Proteica/genética , Análisis de Secuencia de ARN
20.
Transplant Direct ; 1(6): e23, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27500225

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...