Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Front Immunol ; 12: 730545, 2021.
Article in English | MEDLINE | ID: mdl-34566993

ABSTRACT

The human leukocyte antigen G1 (HLA-G1), a non-classical class I major histocompatibility complex (MHC-I) protein, is a potent immunomodulatory molecule at the maternal/fetal interface and other environments to regulate the cellular immune response. We created GGTA1-/HLAG1+ pigs to explore their use as organ and cell donors that may extend xenograft survival and function in both preclinical nonhuman primate (NHP) models and future clinical trials. In the present study, HLA-G1 was expressed from the porcine ROSA26 locus by homology directed repair (HDR) mediated knock-in (KI) with simultaneous deletion of α-1-3-galactotransferase gene (GGTA1; GTKO) using the clustered regularly interspersed palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9) (CRISPR/Cas9) gene-editing system. GTKO/HLAG1+ pigs showing immune inhibitory functions were generated through somatic cell nuclear transfer (SCNT). The presence of HLA-G1 at the ROSA26 locus and the deletion of GGTA1 were confirmed by next generation sequencing (NGS) and Sanger's sequencing. Fibroblasts from piglets, biopsies from transplantable organs, and islets were positive for HLA-G1 expression by confocal microscopy, flow cytometry, or q-PCR. The expression of cell surface HLA-G1 molecule associated with endogenous ß2-microglobulin (ß2m) was confirmed by staining genetically engineered cells with fluorescently labeled recombinant ILT2 protein. Fibroblasts obtained from GTKO/HLAG1+ pigs were shown to modulate the immune response by lowering IFN-γ production by T cells and proliferation of CD4+ and CD8+ T cells, B cells and natural killer (NK) cells, as well as by augmenting phosphorylation of Src homology region 2 domain-containing phosphatase-2 (SHP-2), which plays a central role in immune suppression. Islets isolated from GTKO/HLA-G1+ genetically engineered pigs and transplanted into streptozotocin-diabetic nude mice restored normoglycemia, suggesting that the expression of HLA-G1 did not interfere with their ability to reverse diabetes. The findings presented here suggest that the HLA-G1+ transgene can be stably expressed from the ROSA26 locus of non-fetal maternal tissue at the cell surface. By providing an immunomodulatory signal, expression of HLA-G1+ may extend survival of porcine pancreatic islet and organ xenografts.


Subject(s)
Fibroblasts/metabolism , Galactosyltransferases/deficiency , HLA-G Antigens/metabolism , Killer Cells, Natural/metabolism , T-Lymphocytes/metabolism , Animals , Animals, Genetically Modified , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Blood Glucose/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Fibroblasts/immunology , Galactosyltransferases/genetics , Genotype , HLA-G Antigens/immunology , Haplorhini , Humans , Interferon-gamma/metabolism , Islets of Langerhans Transplantation , Killer Cells, Natural/immunology , Lymphocyte Activation , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Nude , Phenotype , Sus scrofa , T-Lymphocytes/immunology , Tissue Donors , Transplantation, Heterologous
2.
Surg Open Sci ; 4: 26-31, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33937740

ABSTRACT

BACKGROUND: Genetically engineered porcine donors are a potential solution for the shortage of human organs for transplantation. Incompatibilities between humans and porcine donors are largely due to carbohydrate xenoantigens on the surface of porcine cells, provoking an immune response which leads to xenograft rejection. MATERIALS AND METHODS: Multiplex genetic knockout of GGTA1, ß4GalNT2, and CMAH is predicted to increase the rate of xenograft survival, as described previously for GGTA1. In this study, the clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeats-associated protein 9 system was used to target genes relevant to xenotransplantation, and a method for highly efficient editing of multiple genes in primary porcine fibroblasts was described. RESULTS: Editing efficiencies greater than 85% were achieved for knockout of GGTA1, ß4GalNT2, and CMAH. CONCLUSION: The high-efficiency protocol presented here reduces scale and cost while accelerating the production of genetically engineered primary porcine fibroblast cells for in vitro studies and the production of animal models.

3.
HLA ; 96(2): 197-201, 2020 08.
Article in English | MEDLINE | ID: mdl-32452158

ABSTRACT

Pigs are especially useful large animal models, however, limited availability of commercially available antibodies for immunoblotting presents a significant obstacle facing preclinical xenotransplantation research. Major histocompatibility complex class I (MHC-I) molecule expression enhancement by nucleotide-binding oligomerization domain (NOD)-like receptor family with a caspase recruitment domain (CARD) containing caspase 5 (NLRC5) is fundamental to understanding porcine xenoantigen presentation. Swine Leukocyte Antigens (SLAs) are the porcine MHC homologs for human leukocyte antigens. SLA-I is a known xenoantigen that causes T cell activation. NLRC5, SLA-I, and B2M are all targets of immune modulation in genetically engineered pigs in xenotransplantation research with the goal to reduce SLA-I expression. In the present study, the human anti-NLRC5 (ab105411), anti-NLRC5 (ab117624), anti-NLRC5 N-terminal (ab178767), anti-HLA E (ab203082), anti-HLA E (ab135826), anti-HLA E (ab2216) and anti-ß2 M (ab75853) antibodies were examined using immunoblots for porcine cross-reactivity. The anti-human antibodies ab117624, ab105411, ab178767, ab2216, and ab75853 cross reacted with cognate proteins in porcine cell lysates. Antibody reagents from this study will allow for validation of NLRC5, B2M, MHC-I expression in future research studies. In addition, following the methodology described in this study for other xenotransplantation targets may provide an alternative to custom antibody development.


Subject(s)
HLA Antigens , Histocompatibility Antigens Class I , Alleles , Animals , Histocompatibility Antigens Class I/genetics , Humans , Intracellular Signaling Peptides and Proteins , Lymphocyte Activation , Swine
SELECTION OF CITATIONS
SEARCH DETAIL