Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
1.
Front Immunol ; 15: 1418249, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38994362

RESUMEN

Introduction: Decreasing rates of blood donation and close margins between blood supply and demand pose challenges in healthcare. Genetically engineered pig red blood cells (pRBCs) have been explored as alternatives to human RBCs for transfusion, and triple-gene knockout (TKO) modification improves the compatibility of pRBCs with human blood in vitro. In this study, we assessed the efficacy and risks of transfusing wild-type (WT)- and TKO-pRBCs into nonhuman primates (NHPs). Methods: Blood from O-type WT and TKO pigs was processed to produce pRBCs for transfusion, which were transfused or not into NHPs (n=4 per group: WT, TKO, and control) after 25% total blood volume withdrawal: their biological responses were compared. Hematological, biochemical, and immunological parameters were measured before, immediately after, and at intervals following transfusion. Two months later, a second transfusion was performed in three NHPs of the transfusion group. Results: Transfusion of both WT- and TKO-pRBCs significantly improved RBC counts, hematocrit, and hemoglobin levels up to the first day post-transfusion, compared to the controls. The transfusion groups showed instant complement activation and rapid elicitation of anti-pig antibodies, as well as elevated liver enzyme and bilirubin levels post-transfusion. Despite the higher agglutination titers with WT-pRBCs in the pre-transfusion crossmatch, the differences between the WT and TKO groups were not remarkable except for less impairment of liver function in the TKO group. After the second transfusion, more pronounced adverse responses without any hematological gain were observed. Conclusions: WT- and TKO-pRBC transfusions effectively increased hematologic parameters on the first day, with rapid clearance from circulation thereafter. However, pRBC transfusion triggers strong antibody responses, limiting the benefits of the pRBC transfusion and increasing the risk of adverse reactions.


Asunto(s)
Transfusión de Eritrocitos , Eritrocitos , Técnicas de Inactivación de Genes , Animales , Transfusión de Eritrocitos/efectos adversos , Transfusión de Eritrocitos/métodos , Porcinos , Eritrocitos/inmunología , Eritrocitos/metabolismo , Animales Modificados Genéticamente , Hemoglobinas/metabolismo , Galactosiltransferasas/genética , Galactosiltransferasas/deficiencia , Hematócrito , Femenino , Masculino , Primates
2.
Sci Transl Med ; 16(751): eadk6152, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38865482

RESUMEN

Genetic modification of porcine donors, combined with optimized immunosuppression, has been shown to improve outcomes of experimental xenotransplant. However, little is known about outcomes in sensitized recipients, a population that could potentially benefit the most from the clinical implementation of xenotransplantation. Here, five highly allosensitized rhesus macaques received a porcine kidney from GGTA1 (α1,3-galactosyltransferase) knockout pigs expressing the human CD55 transgene (1KO.1TG) and were maintained on an anti-CD154 monoclonal antibody (mAb)-based immunosuppressive regimen. These recipients developed de novo xenoreactive antibodies and experienced xenograft rejection with evidence of thrombotic microangiopathy and antibody-mediated rejection (AMR). In comparison, three highly allosensitized rhesus macaques receiving a kidney from GGTA1, CMAH (cytidine monophospho-N-acetylneuraminic acid hydroxylase), and b4GNT2/b4GALNT2 (ß-1,4-N-acetyl-galactosaminyltransferase 2) knockout pigs expressing seven human transgenes including human CD46, CD55, CD47, THBD (thrombomodulin), PROCR (protein C receptor), TNFAIP3 (tumor necrosis factor-α-induced protein 3), and HMOX1 (heme oxygenase 1) (3KO.7TG) experienced significantly prolonged graft survival and reduced AMR, associated with dampened post-transplant humoral responses, early monocyte and neutrophil activation, and T cell repopulation. After withdrawal of all immunosuppression, recipients who received kidneys from 3KO.7TG pigs rejected the xenografts via AMR. These data suggest that allosensitized recipients may be suitable candidates for xenografts from genetically modified porcine donors and could benefit from an optimized immunosuppression regimen designed to target the post-transplant humoral response, thereby avoiding AMR.


Asunto(s)
Animales Modificados Genéticamente , Galactosiltransferasas , Técnicas de Inactivación de Genes , Rechazo de Injerto , Supervivencia de Injerto , Transgenes , Trasplante Heterólogo , Animales , Supervivencia de Injerto/inmunología , Humanos , Porcinos , Galactosiltransferasas/genética , Galactosiltransferasas/deficiencia , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Macaca mulatta , Trasplante de Riñón
3.
Front Immunol ; 12: 730545, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566993

RESUMEN

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.


Asunto(s)
Fibroblastos/metabolismo , Galactosiltransferasas/deficiencia , Antígenos HLA-G/metabolismo , Células Asesinas Naturales/metabolismo , Linfocitos T/metabolismo , Animales , Animales Modificados Genéticamente , Linfocitos B/inmunología , Linfocitos B/metabolismo , Glucemia/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Fibroblastos/inmunología , Galactosiltransferasas/genética , Genotipo , Antígenos HLA-G/inmunología , Haplorrinos , Humanos , Interferón gamma/metabolismo , Trasplante de Islotes Pancreáticos , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones Desnudos , Fenotipo , Sus scrofa , Linfocitos T/inmunología , Donantes de Tejidos , Trasplante Heterólogo
4.
Eur J Immunol ; 51(6): 1412-1422, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33576494

RESUMEN

Heterologous polyclonal antibodies might represent an alternative to the use of convalescent plasma or monoclonal antibodies (mAbs) in coronavirus disease (COVID-19) by targeting multiple antigen epitopes. However, heterologous antibodies trigger human natural xenogeneic antibody responses particularly directed against animal-type carbohydrates, mainly the N-glycolyl form of the neuraminic acid (Neu5Gc) and the α1,3-galactose, potentially leading to serum sickness or allergy. Here, we immunized cytidine monophosphate-N-acetylneuraminic acid hydroxylase and α1,3-galactosyl-transferase (GGTA1) double KO pigs with the Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike receptor binding domain to produce glyco-humanized polyclonal neutralizing antibodies lacking Neu5Gc and α1,3-galactose epitopes. Animals rapidly developed a hyperimmune response with anti-SARS-CoV-2 end-titers binding dilutions over one to a million and end-titers neutralizing dilutions of 1:10 000. The IgG fraction purified and formulated following clinical Good Manufacturing Practices, named XAV-19, neutralized spike/angiotensin converting enzyme-2 interaction at a concentration <1 µg/mL, and inhibited infection of human cells by SARS-CoV-2 in cytopathic assays. We also found that pig GH-pAb Fc domains fail to interact with human Fc receptors, thereby avoiding macrophage-dependent exacerbated inflammatory responses and a possible antibody-dependent enhancement. These data and the accumulating safety advantages of using GH-pAbs in humans warrant clinical assessment of XAV-19 against COVID-19.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , COVID-19/terapia , SARS-CoV-2/inmunología , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/inmunología , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/farmacología , COVID-19/genética , Galactosiltransferasas/deficiencia , Galactosiltransferasas/inmunología , Células HEK293 , Humanos , Inmunización Pasiva , SARS-CoV-2/genética , Ácidos Siálicos/genética , Ácidos Siálicos/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Porcinos , Sueroterapia para COVID-19
5.
BMC Biotechnol ; 20(1): 40, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811500

RESUMEN

BACKGROUND: Xenoantigens are a major source of concern with regard to the success of interspecific xenografts. GGTA1 encodes α1,3-galactosyltransferase, which is essential for the biosynthesis of galactosyl-alpha 1,3-galactose, the major xenoantigen causing hyperacute rejection. GGTA1-modified pigs, therefore, are promising donors for pig-to-human xenotransplantation. In this study, we developed a method for the introduction of the CRISPR/Cas9 system into in vitro-fertilized porcine zygotes via electroporation to generate GGTA1-modified pigs. RESULTS: We designed five guide RNAs (gRNAs) targeting distinct sites in GGTA1. After the introduction of the Cas9 protein with each gRNA via electroporation, the gene editing efficiency in blastocysts developed from zygotes was evaluated. The gRNA with the highest gene editing efficiency was used to generate GGTA1-edited pigs. Six piglets were delivered from two recipient gilts after the transfer of electroporated zygotes with the Cas9/gRNA complex. Deep sequencing analysis revealed that five out of six piglets carried a biallelic mutation in the targeted region of GGTA1, with no off-target events. Furthermore, staining with isolectin B4 confirmed deficient GGTA1 function in GGTA1 biallelic mutant piglets. CONCLUSIONS: We established GGTA1-modified pigs with high efficiency by introducing a CRISPR/Cas9 system into zygotes via electroporation. Multiple gene modifications, including knock-ins of human genes, in porcine zygotes via electroporation may further improve the application of the technique in pig-to-human xenotransplantation.


Asunto(s)
Sistemas CRISPR-Cas , Electroporación/métodos , Fertilización In Vitro/métodos , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Edición Génica/métodos , Cigoto/metabolismo , Animales , Animales Modificados Genéticamente , Blastocisto , Proteína 9 Asociada a CRISPR/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Disacáridos , Femenino , Xenoinjertos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Mutación , ARN Guía de Kinetoplastida , Porcinos , Trasplante Heterólogo
6.
Lab Anim ; 54(6): 599-604, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32316840

RESUMEN

We report a morphometric evaluation of α1,3-galactosyltransferase knockout (GTKO) pig heart and kidney (n = 9) at the end of one, three and seven months. Organs parameters gradually increased with the age (p < 0.05) and body weight (p < 0.05) of the pigs. Organs morphometries were significantly correlated to the age and body weight of the animal. We were able to conclude the average size of GTKO pig heart and kidney based on age and body weight, which could be helpful in estimating the size of these organs non-invasively for transplantation.


Asunto(s)
Galactosiltransferasas/deficiencia , Corazón/anatomía & histología , Riñón/anatomía & histología , Sus scrofa/anatomía & histología , Animales , Femenino , Técnicas de Inactivación de Genes , Masculino , Sus scrofa/genética
7.
Nat Commun ; 11(1): 860, 2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-32054864

RESUMEN

Glycosylphosphatidylinositol (GPI)-anchored proteins and glycosphingolipids interact with each other in the mammalian plasma membranes, forming dynamic microdomains. How their interaction starts in the cells has been unclear. Here, based on a genome-wide CRISPR-Cas9 genetic screen for genes required for GPI side-chain modification by galactose in the Golgi apparatus, we report that ß1,3-galactosyltransferase 4 (B3GALT4), the previously characterized GM1 ganglioside synthase, additionally functions in transferring galactose to the N-acetylgalactosamine side-chain of GPI. Furthermore, B3GALT4 requires lactosylceramide for the efficient GPI side-chain galactosylation. Thus, our work demonstrates previously unexpected functional relationships between GPI-anchored proteins and glycosphingolipids in the Golgi. Through the same screening, we also show that GPI biosynthesis in the endoplasmic reticulum (ER) is severely suppressed by ER-associated degradation to prevent GPI accumulation when the transfer of synthesized GPI to proteins is defective. Our data demonstrates cross-talks of GPI biosynthesis with glycosphingolipid biosynthesis and the ER quality control system.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Glicoesfingolípidos/biosíntesis , Glicosilfosfatidilinositoles/biosíntesis , Aciltransferasas/deficiencia , Aciltransferasas/genética , Aciltransferasas/metabolismo , Sistemas CRISPR-Cas , Degradación Asociada con el Retículo Endoplásmico/genética , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Galactosiltransferasas/metabolismo , Técnicas de Inactivación de Genes , Glicoesfingolípidos/genética , Glicosilfosfatidilinositoles/genética , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
8.
Res Vet Sci ; 128: 59-68, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31722267

RESUMEN

Handmade cloning is a zona-free nuclear transfer approach and an economical, efficient, and simple micromanipulation-free alternative to dolly based traditional cloning (TC). In this study, based on handmade cloning with minor modifications, an optimized bi-oocyte fusion (BOF) cloning method was established to produce GGTA1 KO porcine embryos using the CRISPR/Cas9 gene editing system. The GGTA1 gene is responsible for the generation of Gal epitopes on the surface of porcine cells, triggering hyperacute immune rejection in preclinical porcine-to-human xenotransplantation. The purpose of the present study is to establish an efficient protocol for activation of porcine oocyte cytoplast-fibroblast fused constructs developed to GGTA1 KO blastocysts by the zona-free bi-oocyte fusion cloning method. High percentages of cleavage (90 ±â€¯2.6%) and blastocyst rates (39 ±â€¯4.0%) were achieved upon treatment with demecolcine-assisted oocyte enucleation followed by 6 V alternating current for proper alignment and single-step fusion technique using a single direct current pulse of 1.0 kV/cm for 9 µs duration, compared to the double-step fusion method with combined chemical activation using thimerosal and dithiothreitol. Overall blastocyst rate was higher for oocyte enucleation by demecolcine (0.4 µg/ml) and 45 min incubation (42 ±â€¯1.5%) compared to without demecolcine incubation followed by complete chemical thimerosal/dithiothreitol activation (33 ±â€¯1.1%). The blastocyst rate (39 ±â€¯1.0%) was found to be significantly higher 1 h post-electrofusion, compared to at 0 and 4 h (28 ±â€¯1.5 and 6 ±â€¯1.5%, respectively). Blastocyst development rates for GGTA1 knockout embryos (38 ±â€¯1.76%) were comparable to those obtained with wild-type embryos (41.1 ±â€¯0.67%). In conclusion, we achieved high overall efficiency in production of GGTA1 KO blastocysts by modified HMC protocol.


Asunto(s)
Animales Modificados Genéticamente , Galactosiltransferasas/metabolismo , Técnicas de Transferencia Nuclear/veterinaria , Sus scrofa , Trasplante Heterólogo/veterinaria , Animales , Sistemas CRISPR-Cas/genética , Galactosiltransferasas/deficiencia , Técnicas de Inactivación de Genes , Oocitos/fisiología
9.
PLoS One ; 14(12): e0226107, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31821359

RESUMEN

The CRISPR/Cas9 gene editing system has enhanced the development of genetically engineered animals for use in xenotransplantation. Potential limitations to the CRISPR/Cas9 system impacting the development of genetically engineered cells and animals include the creation of off-target mutations. We sought to develop a method to reduce the likelihood of off-target mutation while maintaining a high efficiency rate of desired genetic mutations for the GGTA1 gene. Extension of sgRNA length, responsible for recognition of the target DNA sequence for Cas9 cleavage, resulted in improved specificity for the GGTA1 gene and less off-target DNA cleavage. Three PAM sites were selected within exon 1 of the porcine GGTA1 gene and ten sgRNA of variable lengths were designed across these three sites. The sgRNA was tested against synthetic double stranded DNA templates replicating both the native GGTA1 DNA template and the two most likely off-target binding sites in the porcine genome. Cleavage ability for native and off-target DNA was determined by in vitro cleavage assays. Resulting cleavage products were analyzed to determine the cleavage efficiency of the Cas9/sgRNA complex. Extension of sgRNA length did not have a statistical impact on the specificity of the Cas9/sgRNA complex for PAM1 and PAM2 sites. At the PAM3 site, however, an observed increase in specificity for native versus off-target templates was seen with increased sgRNA length. In addition, distance between PAM site and the start codon had a significant impact on cleavage efficiency and target specificity, regardless of sgRNA length. Although the in vitro assays showed off-target cleavage, Sanger sequencing revealed that no off-target mutations were found in GGTA1 knockout cell lines or piglet. These results demonstrate an optimized method for improvement of the CRIPSR/Cas9 gene editing system by reducing the likelihood of damaging off-target mutations in GGTA1 knocked out cells destined for xenotransplant donor production.


Asunto(s)
Sistemas CRISPR-Cas/genética , ADN/metabolismo , Galactosiltransferasas/genética , Edición Génica/métodos , ARN Guía de Kinetoplastida/metabolismo , Animales , Sitios de Unión , ADN/genética , División del ADN , Galactosiltransferasas/deficiencia , ARN Guía de Kinetoplastida/química , Ribonucleoproteínas/metabolismo , Porcinos
10.
Sci Rep ; 9(1): 7133, 2019 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-31073169

RESUMEN

The role of carbohydrate chains in leukocyte migration to inflamed sites during inflammation and trafficking to the lymph nodes under physiological conditions has been extensively characterized. Here, we report that carbohydrate chains also mediate the homing and engraftment of hematopoietic stem/progenitor cells (HSPCs) to the bone marrow (BM). In particular, we found that transplanted BM cells deficient in ß-1,4-galactosyltransferase-1 (ß4GalT-1) could not support survival in mice exposed to a lethal dose of irradiation. BM cells obtained from mice deficient in ß4GalT-1 showed normal colony-forming activity and hematopoietic stem cell numbers. However, colony-forming cells were markedly rare in the BM of recipient mice 24 h after transplantation of ß4GalT-1-deficient BM cells, suggesting that ß4GalT-1 deficiency severely impairs homing. Similarly, BM cells with a point mutation in the UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase gene, encoding a key enzyme in sialic acid biosynthesis, showed mildly impaired homing and engraftment abilities. These results imply that the galactosyl, but not sialyl residues in glycoproteins, are essential for the homing and engraftment of HSPCs to the BM. These findings suggest the possibility of modifying carbohydrate structures on the surface of HSPCs to improve their homing and engraftment to the BM in clinical application.


Asunto(s)
Células de la Médula Ósea/citología , Galactosiltransferasas/deficiencia , Células Madre Hematopoyéticas/citología , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Metabolismo de los Hidratos de Carbono , Células Cultivadas , Femenino , Galactosiltransferasas/genética , Ratones , Mutación Puntual
11.
Dis Model Mech ; 12(6)2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31101663

RESUMEN

In a screen for organogenesis defects in N-ethyl-N-nitrosourea (ENU)-induced mutant mice, we discovered a line carrying a mutation in Colgalt1 [collagen beta(1-O)galactosyltransferase type 1], which is required for proper galactosylation of hydroxylysine residues in a number of collagens. Colgalt1 mutant embryos have not been previously characterized; here, we show that they exhibit skeletal and muscular defects. Analysis of mutant-derived embryonic fibroblasts reveals that COLGALT1 acts on collagen IV and VI, and, while collagen VI appears stable and its secretion is not affected, collagen IV accumulates inside of cells and within the extracellular matrix, possibly due to instability and increased degradation. We also generated mutant zebrafish that do not express the duplicated orthologs of mammalian Colgalt1 The double-homozygote mutants have muscle defects; they are viable through the larvae stage but do not survive to 10 days post-fertilization. We hypothesize that the Colgalt1 mutant could serve as a model of a human connective tissue disorder and/or congenital muscular dystrophy or myopathy.


Asunto(s)
Colágeno/metabolismo , Galactosiltransferasas/deficiencia , Mutación con Pérdida de Función/genética , Sistema Musculoesquelético/patología , Procesamiento Proteico-Postraduccional , Proteínas de Pez Cebra/metabolismo , Alelos , Animales , Embrión de Mamíferos/patología , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Galactosiltransferasas/metabolismo , Glicosilación , Ratones , Peso Molecular , Músculos/metabolismo , Músculos/patología , Mutación Missense/genética , Fenotipo , Piel/metabolismo , Piel/patología , Pez Cebra
12.
Xenotransplantation ; 26(5): e12524, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31115108

RESUMEN

Two well-characterized carbohydrate epitopes are absent in humans but present in other mammals. These are galactose-α1,3-galactose (αGal) and N-glycolylneuraminic acid (Neu5Gc) which are introduced by the activities of two enzymes including α(1,3) galactosyltransferase (encoded by the GGTA1 gene) and CMP-Neu5Gc hydroxylase (encoded by the CMAH gene) that are inactive in humans but present in cattle. Hence, bovine-derived products are antigenic in humans who receive bioprosthetic heart valves (BHVs) or those that suffer from red meat syndrome. Using programmable nucleases, we disrupted (knockout, KO) GGTA1 and CMAH genes encoding for the enzymes that catalyse the synthesis of αGal and Neu5Gc, respectively, in both male and female bovine fibroblasts. The KO in clonally selected fibroblasts was detected by polymerase chain reaction (PCR) and confirmed by Sanger sequencing. Selected fibroblasts colonies were used for somatic cell nuclear transfer (SCNT) to produce cloned embryos that were implanted in surrogate recipient heifers. Fifty-three embryos were implanted in 33 recipients heifers; 3 pregnancies were carried to term and delivered 3 live calves. Primary cell cultures were established from the 3 calves and following molecular analyses confirmed the genetic deletions. FACS analysis showed the double-KO phenotype for both antigens confirming the mutated genotypes. Availability of such cattle double-KO model lacking both αGal and Neu5Gc offers a unique opportunity to study the functionality of BHV manufactured with tissues of potentially lower immunogenicity, as well as a possible new clinical approaches to help patients with red meat allergy syndrome due to the presence of these xenoantigens in the diet.


Asunto(s)
Animales Modificados Genéticamente , Antígenos Heterófilos/metabolismo , Citidina Monofosfato/análogos & derivados , Galactosa/metabolismo , Galactosiltransferasas/genética , Técnicas de Inactivación de Genes , Oxigenasas de Función Mixta/genética , Ácidos Neuramínicos/metabolismo , Animales , Antígenos Heterófilos/inmunología , Bioprótesis , Bovinos , Citidina Monofosfato/inmunología , Citidina Monofosfato/metabolismo , Femenino , Fibroblastos/inmunología , Hipersensibilidad a los Alimentos/inmunología , Galactosa/inmunología , Galactosiltransferasas/deficiencia , Prótesis Valvulares Cardíacas , Humanos , Masculino , Oxigenasas de Función Mixta/deficiencia , Ácidos Neuramínicos/inmunología , Trasplante Heterólogo
13.
Xenotransplantation ; 26(4): e12516, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30989742

RESUMEN

Xenotransplantation research has made considerable progress in recent years, largely through the increasing availability of pigs with multiple genetic modifications. We suggest that a pig with nine genetic modifications (ie, currently available) will provide organs (initially kidneys and hearts) that would function for a clinically valuable period of time, for example, >12 months, after transplantation into patients with end-stage organ failure. The national regulatory authorities, however, will likely require evidence, based on in vitro and/or in vivo experimental data, to justify the inclusion of each individual genetic modification in the pig. We provide data both from our own experience and that of others on the advantages of pigs in which (a) all three known carbohydrate xenoantigens have been deleted (triple-knockout pigs), (b) two human complement-regulatory proteins (CD46, CD55) and two human coagulation-regulatory proteins (thrombomodulin, endothelial cell protein C receptor) are expressed, (c) the anti-apoptotic and "anti-inflammatory" molecule, human hemeoxygenase-1 is expressed, and (d) human CD47 is expressed to suppress elements of the macrophage and T-cell responses. Although many alternative genetic modifications could be made to an organ-source pig, we suggest that the genetic manipulations we identify above will all contribute to the success of the initial clinical pig kidney or heart transplants, and that the beneficial contribution of each individual manipulation is supported by considerable experimental evidence.


Asunto(s)
Animales Modificados Genéticamente/genética , Rechazo de Injerto/prevención & control , Porcinos/genética , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente/inmunología , Antígeno CD47/genética , Antígeno CD47/inmunología , Antígenos CD55/genética , Antígenos CD55/inmunología , Receptor de Proteína C Endotelial/genética , Receptor de Proteína C Endotelial/inmunología , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Galactosiltransferasas/inmunología , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Rechazo de Injerto/inmunología , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/inmunología , Humanos , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/inmunología , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/inmunología , N-Acetilgalactosaminiltransferasas/deficiencia , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/inmunología , Porcinos/inmunología , Trombomodulina/genética , Trombomodulina/inmunología
14.
Bone ; 124: 14-21, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30914273

RESUMEN

B4GALT7 encodes beta-1,4-galactosyltransferase which links glycosaminoglycans to proteoglycans in connective tissues. Rare, biallelic variants in B4GALT7 have been associated with spondylodysplastic Ehlers-Danlos and Larsen of Reunion Island syndromes. Thirty patients with B4GALT7-related disorders have been reported to date with phenotypic variability. Using whole exome sequencing, we identified male and female siblings with biallelic, pathogenic B4GALT7 variants and phenotypic features of spondylodysplastic Ehlers-Danlos syndrome as well as previously unreported skeletal characteristics. We also provide detailed radiological characterization and describe the siblings' responses to growth hormone treatment. Our report extends the phenotypic spectrum of B4GALT7-associated spondylodysplastic Ehlers-Danlos syndrome and reports results of growth hormone treatment for patients with this rare disorder.


Asunto(s)
Galactosiltransferasas/deficiencia , Hormona del Crecimiento/uso terapéutico , Hermanos , Adulto , Biomarcadores/metabolismo , Niño , Preescolar , Femenino , Galactosiltransferasas/genética , Galactosiltransferasas/metabolismo , Humanos , Masculino , Fenotipo , Secuenciación del Exoma
15.
Xenotransplantation ; 26(4): e12504, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30825348

RESUMEN

The humoral barrier has been the limiting factor in moving xenotransplantation towards the clinic. Improvements in somatic cell nuclear transfer and genome editing, particularly CRISPR-Cas9, have made it possible to create pigs with multiple glycan xenoantigen deletions for the purposes of reducing xenoreactive antibody binding to the xenografted organ. Recent studies have also considered the aetiology and existence of antibodies directed at the swine leucocyte antigen (SLA) complex, and potential genetic engineering strategies to avoid these antibodies. Evaluation of xenoreactive antibody binding is very important for the advancement of xenotransplantation, because if patients do not have any detectable xenoreactive antibody, then it is reasonable to expect that cellular rejection and not antibody-mediated rejection (AMR) will be the next hurdle to clinical application.


Asunto(s)
Antígenos Heterófilos/inmunología , Galactosiltransferasas/inmunología , Técnicas de Inactivación de Genes , Rechazo de Injerto/prevención & control , Oxigenasas de Función Mixta/inmunología , N-Acetilgalactosaminiltransferasas/inmunología , Porcinos/inmunología , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente/inmunología , Anticuerpos Heterófilos/biosíntesis , Anticuerpos Heterófilos/inmunología , Reacciones Antígeno-Anticuerpo , Antígenos Heterófilos/genética , Epítopos/inmunología , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Ingeniería Genética , Rechazo de Injerto/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , N-Acetilgalactosaminiltransferasas/deficiencia , N-Acetilgalactosaminiltransferasas/genética , Inmunología del Trasplante
16.
Nature ; 564(7736): 430-433, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30518863

RESUMEN

Heart transplantation is the only cure for patients with terminal cardiac failure, but the supply of allogeneic donor organs falls far short of the clinical need1-3. Xenotransplantation of genetically modified pig hearts has been discussed as a potential alternative4. Genetically multi-modified pig hearts that lack galactose-α1,3-galactose epitopes (α1,3-galactosyltransferase knockout) and express a human membrane cofactor protein (CD46) and human thrombomodulin have survived for up to 945 days after heterotopic abdominal transplantation in baboons5. This model demonstrated long-term acceptance of discordant xenografts with safe immunosuppression but did not predict their life-supporting function. Despite 25 years of extensive research, the maximum survival of a baboon after heart replacement with a porcine xenograft was only 57 days and this was achieved, to our knowledge, only once6. Here we show that α1,3-galactosyltransferase-knockout pig hearts that express human CD46 and thrombomodulin require non-ischaemic preservation with continuous perfusion and control of post-transplantation growth to ensure long-term orthotopic function of the xenograft in baboons, the most stringent preclinical xenotransplantation model. Consistent life-supporting function of xenografted hearts for up to 195 days is a milestone on the way to clinical cardiac xenotransplantation7.


Asunto(s)
Trasplante de Corazón , Xenoinjertos/trasplante , Papio , Porcinos , Trasplante Heterólogo , Animales , Anticuerpos/análisis , Anticuerpos/sangre , Proteínas del Sistema Complemento/análisis , Enzimas/sangre , Fibrina/análisis , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Xenoinjertos/patología , Humanos , Hígado/enzimología , Masculino , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/metabolismo , Miocardio/enzimología , Necrosis , Perfusión , Recuento de Plaquetas , Tiempo de Protrombina , Trombomodulina/genética , Trombomodulina/metabolismo , Factores de Tiempo
17.
PLoS One ; 13(11): e0207272, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30462716

RESUMEN

The liver's regenerative capacity is unique, but too small a segment can overwhelm its ability to simultaneously regenerate and support the host, resulting in liver dysfunction and death. Here we tested a temporary Xenogeneic Heterotopic Auxiliary Liver Transplant (XHALT) from Gal-KO miniature swine in a baboon model of Post-Hepatectomy Liver Failure (PHLF) by 90%- hepatectomy. Immunosuppression consisted of CVF, ATG, FK 506 and steroids. 90%-hepatectomized animals died within 4-5 days with the clinical picture of PHLF, (high LFTs and bilirubin, ascites, encephalopathy and coagulopathy). The 10% remnants had macroscopic and histological evidence of severe steatosis and absence of hepatocyte replication. In contrast, the addition of XHALT prolonged survival up to 11 days, with the cause of death being sepsis, rather than liver failure. The remnant liver appeared grossly normal, and on histology, there was no evidence of fatty infiltration, but there was pronounced Ki-67 staining. In conclusion, temporary auxiliary xenografts have the potential to support a small for size liver graft while it grows to adequate size or provide an opportunity for organ recovery in acute liver failure.


Asunto(s)
Fallo Hepático/cirugía , Regeneración Hepática/fisiología , Trasplante de Hígado/métodos , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Técnicas de Inactivación de Genes , Supervivencia de Injerto , Hepatectomía , Xenoinjertos , Fallo Hepático/patología , Fallo Hepático/fisiopatología , Papio , Porcinos , Porcinos Enanos , Trasplante Heterotópico
18.
Cell Physiol Biochem ; 50(3): 1186-1200, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30355948

RESUMEN

BACKGROUND/AIMS: The elaborate structure of the extracellular matrix (ECM) and the appropriate surface glycoforms upon it are indispensable to CD4+ T cell regulation. METHODS: To explore the effects of Glcα1,2Galß1 glycosylation mediated by GLT25D2 (Colgalt2) for CD4+ T cell regulation, we prepared C57BL/6J Glt25d2-/- mice. In the induction of hepatitis, after concanavalin A (Con A) challenge for 6, 12, and 24 h, more extensive parenchymal injury was noted in Glt25d2-/- mice than in wild-type (WT) mice at 12 h. Immunohistochemistry and laser scanning confocal microscopy were used to detect GLT25D2 expression, and subsets of CD4+T cells was analyzed by flow cytometry. A total of 26 cytokines in serum samples were determined using Luminex technology. RESULTS: The trend in liver injury score variation was consistent with serum alanine aminotransferase and aspartate aminotransferase levels. The levels of interleukin 4 (IL-4), IL-1ß, IL-9, and several chemokines such as macrophage inflammatory protein-2, eotaxin, and growth-related oncogene α were significantly increased in Glt25d2-/- mice compared with WT mice after Con A challenge. A further phenotype analysis of primary Glt25d2-/- CD4+ T cells showed that Glt25d2 knockout increased the frequency of the CD25+CD69- subset but decreased the frequency of the CD25-CD69+ subset after Con A challenge for 6, 12, and 24 h compared with those of WT CD4+ T cells. Activation-induced apoptosis was also significantly increased in Glt25d2-/- CD4+ T cells after Con A challenge compared with WT CD4+ T cells. Lectin microarray hybridization showed that Glt25d2 knockout increased the binding activity of Narcissus pseudonarcissus lectin to CD4+ T cells but Amaranthus caudatus lectin-binding activity was lost during Con A challenge. CONCLUSION: The present results suggest that collagen glycosylation mediated by GLT25D2 may regulate a subset of CD4+ T cells and be involved in the pathogenesis of Con A-induced hepatitis.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Concanavalina A/farmacología , Galactosiltransferasas/genética , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Apoptosis/efectos de los fármacos , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Quimiocinas/sangre , Citocinas/sangre , Galactosiltransferasas/deficiencia , Hepatitis Animal/etiología , Hepatitis Animal/inmunología , Hepatitis Animal/patología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Lectinas/metabolismo , Lectinas Tipo C/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/metabolismo
19.
Artif Cells Nanomed Biotechnol ; 46(sup3): S359-S369, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30207744

RESUMEN

Homeotransplantation of bones for replacement therapy have been demonstrated reliably in clinical data. However, human donor bones applicable for homeotransplantation are in short supply, which facilitates the search for suitable alternatives, such as xenografts grafts. The α-Gal antigen-related immune risk of xenografts directly affects the safety and effectiveness of the biomaterials and limits their applications in the clinic. The immune risk can be prevented by depletion or breaking anti-Gal antibody prior to transplant. Therefore, how to assess the immune risk of the bone substitutes and select the reliable animal research model become extremely important. In this study, we prepared lyophilized bone substitutes (T1) and Guanghao Biotech bone substitutes (T2, animal-derived biomaterials with α-Gal antigen decreased), aimed to assess the immune risk of xenografts bone substitutes on GGTA1 knockout mice. The α-Gal antigen contents of T1 and T2 were firstly detected by ELISA method in vitro. The bone substitutes were then implanted subcutaneously into GGTA1 knockout mice for 2, 4 and 12 weeks, respectively. The total serum antibody levels, anti-α-Gal antibody levels, inflammatory cytokine and splenic lymphocyte surface molecules were detected and histology analysis of skin and thymus were performed to systematically evaluate the immune response caused by the T1 and T2 bone substitutes in mice. In vitro results showed that the amount of α-Gal epitopes in T1 bone substitutes was significantly higher than T2 bone substitutes, and the clearance rate of α-Gal antigen in T2 bone substitutes achieved about 55.6%. Results of antibody level in vivo showed that the T1 bone substitutes group possessed significantly higher total IgG, IgM, IgA and anti-α-Gal IgG levels than T2 and control group, while T2 group showed no significant changes of these indexes compared with control. In terms of inflammatory cytokines, T1 bone substitutes showed evidently higher levels of IL-4, IL-12P70 and IL-10 than T2 and control, while T2 group was comparable to control. No changes in the levels of splenic lymphocyte surface molecules were found in the three groups (T1, T2 and control group) during the experimental periods. The pathological results demonstrated that the inflammatory response in T2 group was lighter than the T1 group, which was in accordance with the inflammatory cytokines levels. The above results indicated that the process of antigen removal effectively reduced the α-Gal antigens content in T2 bone substitutes, which caused little immune response in vivo and could be used as bone healing materials. This study also demonstrated that GGTA1 knockout mice can be used as a routine tool to assess the immune risk of animal-derived biomaterials.


Asunto(s)
Matriz Ósea , Trasplante Óseo , Galactosiltransferasas/deficiencia , Animales , Anticuerpos/inmunología , Matriz Ósea/inmunología , Matriz Ósea/patología , Matriz Ósea/trasplante , Sustitutos de Huesos/farmacología , Galactosiltransferasas/inmunología , Xenoinjertos , Ratones , Ratones Noqueados , Trasplante Heterólogo
20.
Mol Biotechnol ; 60(8): 550-562, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29916131

RESUMEN

Pig-to-human organ transplantation has drawn attention in recent years due to the potential use of pigs as an alternative source of human donor organs. While GGTA1 knockout (GTKO) can protect xenografts from hyperacute rejection, complement-dependent cytotoxicity might still contribute to this type of rejection. To prolong the xenograft survival, we utilized a T2A-mediated pCMV-hCD55-T2A-hCD59-Neo vector and transfected the plasmid into GTKO Diannan miniature pig fetal fibroblasts. After G418 selection combined with single-cell cloning culture, four colonies were obtained, and three of these were successfully transfected with the hCD55 and hCD59. One of the three colonies was selected as donor cells for somatic cell nuclear transfer (SCNT). Then, the reconstructed embryos were transferred into eight recipient gilts, resulting in four pregnancies. Three of the pregnant gilts delivered, yielding six piglets. Only one piglet carried hCD55 and hCD59 genetic modification. The expression levels of the GGTA1, hCD55, and hCD59 in the tissues and fibroblasts of the piglet were determined by q-PCR, fluorescence microscopy, immunohistochemical staining, and western blotting analyses. The results showed the absence of GGTA1 and the coexpression of the hCD55 and hCD59. However, the mRNA expression levels of hCD55 and hCD59 in the GTKO/hCD55/hCD59 pig fibroblasts were lower than that in human 293T cells, which may be caused by low copy number and/or CMV promoter methylation. Furthermore, we performed human complement-mediated cytolysis assays using human serum solutions from 0 to 60%. The result showed that the fibroblasts of this triple-gene modified piglet had greater survival rates than that of wild-type and GTKO controls. Taken together, these results indicate that T2A-mediated polycistronic vector system combined with SCNT can effectively generate multiplex genetically modified pigs, additional hCD55 and hCD59 expression on top of a GTKO genetic background markedly enhance the protective effect towards human serum-mediated cytolysis than those of GTKO alone. Thus, we suggest that GTKO/hCD55/hCD59 triple-gene-modified Diannan miniature pig will be a more eligible donor for xenotransplantation.


Asunto(s)
Antígenos CD55/genética , Antígenos CD59/genética , Galactosiltransferasas/deficiencia , Vectores Genéticos/genética , Técnicas de Transferencia Nuclear , Péptidos/genética , Animales , Animales Modificados Genéticamente , Antígenos CD55/inmunología , Antígenos CD55/metabolismo , Antígenos CD59/inmunología , Antígenos CD59/metabolismo , Ensayo de Actividad Hemolítica de Complemento , Femenino , Fibroblastos/metabolismo , Galactosiltransferasas/metabolismo , Expresión Génica , Humanos , Péptidos/química , Embarazo , ARN Mensajero/metabolismo , Porcinos , Porcinos Enanos/genética , Distribución Tisular , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...