Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Lab Invest ; 102(5): 560-569, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34980882

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease, manifesting as the progressive development of fluid-filled renal cysts. In approximately half of all patients with ADPKD, end-stage renal disease results in decreased renal function. In this study, we used CRISPR-Cas9 and somatic cell cloning to produce pigs with the unique mutation c.152_153insG (PKD1insG/+). Pathological analysis of founder cloned animals and progeny revealed that PKD1insG/+ pigs developed many pathological conditions similar to those of patients with heterozygous mutations in PKD1. Pathological similarities included the formation of macroscopic renal cysts at the neonatal stage, number and cystogenic dynamics of the renal cysts formed, interstitial fibrosis of the renal tissue, and presence of a premature asymptomatic stage. Our findings demonstrate that PKD1insG/+ pigs recapitulate the characteristic symptoms of ADPKD.


Asunto(s)
Riñón Poliquístico Autosómico Dominante , Animales , Femenino , Heterocigoto , Humanos , Riñón/patología , Masculino , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Porcinos , Canales Catiónicos TRPP/genética
2.
J Reprod Dev ; 62(5): 511-520, 2016 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-27396383

RESUMEN

Genetically modified pigs that express fluorescent proteins such as green and red fluorescent proteins have become indispensable biomedical research tools in recent years. Cell or tissue transplantation studies using fluorescent markers should be conducted, wherein the xeno-antigenicity of the fluorescent proteins does not affect engraftment or graft survival. Thus, we aimed to create a transgenic (Tg)-cloned pig that was immunologically tolerant to fluorescent protein antigens. In the present study, we generated a Tg-cloned pig harboring a derivative of Plum modified by a single amino acid substitution in the chromophore. The cells and tissues of this Tg-cloned pig expressing the modified Plum (mPlum) did not fluoresce. However, western blot and immunohistochemistry analyses clearly showed that the mPlum had the same antigenicity as Plum. Thus, we have obtained primary proof of principle for creating a cloned pig that is immunologically tolerant to fluorescent protein antigens.


Asunto(s)
Animales Modificados Genéticamente , Técnicas de Transferencia Nuclear , Transgenes , Animales , Antígenos/metabolismo , Núcleo Celular/metabolismo , Clonación Molecular , Metilación de ADN , Femenino , Fibroblastos/metabolismo , Citometría de Flujo , Fluorescencia , Vectores Genéticos , Genotipo , Supervivencia de Injerto , Inmunohistoquímica , Sustancias Luminiscentes/química , Porcinos
3.
J Reprod Dev ; 61(3): 169-77, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25739316

RESUMEN

Monomeric Plum (Plum), a far-red fluorescent protein with photostability and photopermeability, is potentially suitable for in vivo imaging and detection of fluorescence in body tissues. The aim of this study was to generate transgenic cloned pigs exhibiting systemic expression of Plum using somatic cell nuclear transfer (SCNT) technology. Nuclear donor cells for SCNT were obtained by introducing a Plum-expression vector driven by a combination of the cytomegalovirus early enhancer and chicken beta-actin promoter into porcine fetal fibroblasts (PFFs). The cleavage and blastocyst formation rates of reconstructed SCNT embryos were 81.0% (34/42) and 78.6% (33/42), respectively. At 36-37 days of gestation, three fetuses systemically expressing Plum were obtained from one recipient to which 103 SCNT embryos were transferred (3/103, 2.9%). For generation of offspring expressing Plum, rejuvenated PFFs were established from one cloned fetus and used as nuclear donor cells. Four cloned offspring and one stillborn cloned offspring were produced from one recipient to which 117 SCNT embryos were transferred (5/117, 4.3%). All offspring exhibited high levels of Plum fluorescence in blood cells, such as lymphocytes, monocytes and granulocytes. In addition, the skin, heart, kidney, pancreas, liver and spleen also exhibited Plum expression. These observations demonstrated that transfer of the Plum gene did not interfere with the development of porcine SCNT embryos and resulted in the successful generation of transgenic cloned pigs that systemically expressed Plum. This is the first report of the generation and characterization of transgenic cloned pigs expressing the far-red fluorescent protein Plum.


Asunto(s)
Animales Modificados Genéticamente , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/química , Técnicas de Transferencia Nuclear , Actinas/metabolismo , Animales , Blastocisto/citología , Núcleo Celular/metabolismo , Pollos , Clonación de Organismos , Femenino , Fibroblastos/metabolismo , Vectores Genéticos , Granulocitos/citología , Linfocitos/citología , Monocitos/citología , Regiones Promotoras Genéticas , Porcinos , Proteína Fluorescente Roja
4.
J Reprod Dev ; 61(5): 449-57, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26227017

RESUMEN

Zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) are new tools for producing gene knockout (KO) animals. The current study reports produced genetically modified pigs, in which two endogenous genes were knocked out. Porcine fibroblast cell lines were derived from homozygous α1,3-galactosyltransferase (GalT) KO pigs. These cells were subjected to an additional KO for the cytidine monophospho-N-acetylneuraminic acid hydroxylase (CMAH) gene. A pair of ZFN-encoding mRNAs targeting exon 8 of the CMAH gene was used to generate the heterozygous CMAH KO cells, from which cloned pigs were produced by somatic cell nuclear transfer (SCNT). One of the cloned pigs obtained was re-cloned after additional KO of the remaining CMAH allele using the same ZFN-encoding mRNAs to generate GalT/CMAH-double homozygous KO pigs. On the other hand, the use of TALEN-encoding mRNAs targeting exon 7 of the CMAH gene resulted in efficient generation of homozygous CMAH KO cells. These cells were used for SCNT to produce cloned pigs homozygous for a double GalT/CMAH KO. These results demonstrate that the combination of TALEN-encoding mRNA, in vitro selection of the nuclear donor cells and SCNT provides a robust method for generating KO pigs.


Asunto(s)
Animales Modificados Genéticamente/genética , Galactosiltransferasas/genética , Técnicas de Inactivación de Genes/veterinaria , Oxigenasas de Función Mixta/genética , Sus scrofa/genética , Alelos , Animales , Animales Modificados Genéticamente/metabolismo , Animales Recién Nacidos , Línea Celular , Clonación de Organismos/veterinaria , Transferencia de Embrión/veterinaria , Exones , Femenino , Galactosiltransferasas/antagonistas & inhibidores , Galactosiltransferasas/metabolismo , Homocigoto , Japón , Masculino , Oxigenasas de Función Mixta/antagonistas & inhibidores , Oxigenasas de Función Mixta/metabolismo , Técnicas de Transferencia Nuclear/veterinaria , ARN/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Sus scrofa/metabolismo
5.
Regen Ther ; 24: 451-458, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37772130

RESUMEN

Introduction: Duchenne muscular dystrophy (DMD) is a hereditary neuromuscular disorder caused by mutation in the dystrophin gene (DMD) on the X chromosome. Female DMD carriers occasionally exhibit symptoms such as muscle weakness and heart failure. Here, we investigated the characteristics and representativeness of female DMD carrier (DMD-XKOXWT) pigs as a suitable disease model. Methods: In vitro fertilization using sperm from a DMD-XKOY↔XWTXWT chimeric boar yielded DMD-XKOXWT females, which were used to generate F2 and F3 progeny, including DMD-XKOXWT females. F1-F3 piglets were genotyped and subjected to biochemical analysis for blood creatine kinase (CK), aspartate aminotransferase, and lactate dehydrogenase. Skeletal muscle and myocardial tissue were analyzed for the expression of dystrophin and utrophin, as well as for lymphocyte and macrophage infiltration. Results: DMD-XKOXWT pigs exhibited various characteristics common to human DMD carrier patients, namely, asymptomatic hyperCKemia, dystrophin expression patterns in the skeletal and cardiac muscles, histopathological features of skeletal muscle degeneration, myocardial lesions in adulthood, and sporadic death. Pathological abnormalities observed in the skeletal muscles in DMD-XKOXWT pigs point to a frequent incidence of pathological abnormalities in the musculoskeletal tissues of latent DMD carriers. Our findings suggest a higher risk of myocardial abnormalities in DMD carrier women than previously believed. Conclusions: We demonstrated that DMD-XKOXWT pigs could serve as a suitable large animal model for understanding the pathogenic mechanism in DMD carriers and developing therapies for female DMD carriers.

6.
Mol Ther Nucleic Acids ; 33: 444-453, 2023 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-37588685

RESUMEN

Mammalian artificial chromosomes have enabled the introduction of extremely large amounts of genetic information into animal cells in an autonomously replicating, nonintegrating format. However, the evaluation of human artificial chromosomes (HACs) as novel tools for curing intractable hereditary disorders has been hindered by the limited efficacy of the delivery system. We generated dystrophin gene knockout (DMD-KO) pigs harboring the HAC bearing the entire human DMD via a somatic cell cloning procedure (DYS-HAC-cloned pig). Restored human dystrophin expression was confirmed by immunofluorescence staining in the skeletal muscle of the DYS-HAC-cloned pigs. Viability at the first month postpartum of the DYS-HAC-cloned pigs, including motor function in the hind leg and serum creatinine kinase level, was improved significantly when compared with that in the original DMD-KO pigs. However, decrease in systemic retention of the DYS-HAC vector and limited production of the DMD protein might have caused severe respiratory impairment with general prostration by 3 months postpartum. The results demonstrate that the use of transchromosomic cloned pigs permitted a straightforward estimation of the efficacy of the DYS-HAC carried in affected tissues/organs in a large-animal disease model, providing novel insights into the therapeutic application of exogenous mammalian artificial chromosomes.

7.
Biochem Biophys Res Commun ; 402(1): 14-8, 2010 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-20875794

RESUMEN

Zinc-finger nucleases (ZFNs) are expected as a powerful tool for generating gene knockouts in laboratory and domestic animals. Currently, it is unclear whether this technology can be utilized for knocking-out genes in pigs. Here, we investigated whether knockout (KO) events in which ZFNs recognize and cleave a target sequence occur in porcine primary cultured somatic cells that harbor the exogenous enhanced green fluorescent protein (EGFP) gene. ZFN-encoding mRNA designed to target the EGFP gene was introduced by electroporation into the cell. Using the Surveyor nuclease assay and flow cytometric analysis, we confirmed ZFN-induced cleavage of the target sequence and the disappearance of EGFP fluorescence expression in ZFN-treated cells. In addition, sequence analysis revealed that ZFN-induced mutations such as base substitution, deletion, or insertion were generated in the ZFN cleavage site of EGFP-expression negative cells that were cloned from ZFN-treated cells, thereby showing it was possible to disrupt (i.e., knock out) the function of the EGFP gene in porcine somatic cells. To our knowledge, this study provides the first evidence that the ZFN-KO system can be applied to pigs. These findings may open a new avenue to the creation of gene KO pigs using ZFN-treated cells and somatic cell nuclear transfer.


Asunto(s)
Animales Modificados Genéticamente/genética , Desoxirribonucleasas/metabolismo , Técnicas de Inactivación de Genes/métodos , Proteínas Fluorescentes Verdes/genética , Sus scrofa/genética , Dedos de Zinc , Animales , Secuencia de Bases , Células Cultivadas , División del ADN , Análisis Mutacional de ADN , Desoxirribonucleasas/genética , Electroporación , Datos de Secuencia Molecular
8.
Sci Rep ; 9(1): 8016, 2019 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-31142767

RESUMEN

To combat organ shortage in transplantation medicine, a novel strategy has been proposed to generate human organs from exogenous pluripotent stem cells utilizing the developmental mechanisms of pig embryos/foetuses. Genetically modified pigs missing specific organs are key elements in this strategy. In this study, we demonstrate the feasibility of using a genome-editing approach to generate anephrogenic foetuses in a genetically engineered pig model. SALL1 knockout (KO) was successfully induced by injecting genome-editing molecules into the cytoplasm of pig zygotes, which generated the anephrogenic phenotype. Extinguished SALL1 expression and marked dysgenesis of nephron structures were observed in the rudimentary kidney tissue of SALL1-KO foetuses. Biallelic KO mutations of the target gene induced nephrogenic defects; however, biallelic mutations involving small in-frame deletions did not induce the anephrogenic phenotype. Through production of F1 progeny from mutant founder pigs, we identified mutations that could reliably induce the anephrogenic phenotype and hence established a line of fertile SALL1-mutant pigs. Our study lays important technical groundwork for the realization of human kidney regeneration through the use of an empty developmental niche in pig foetuses.


Asunto(s)
Animales Modificados Genéticamente , Edición Génica/métodos , Nefronas/crecimiento & desarrollo , Ingeniería de Tejidos/métodos , Factores de Transcripción/genética , Aloinjertos/provisión & distribución , Animales , Sistemas CRISPR-Cas/genética , Estudios de Factibilidad , Femenino , Desarrollo Fetal/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Trasplante de Riñón , Masculino , Mutación , Células Madre Pluripotentes/fisiología , Regeneración/fisiología , Sus scrofa , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética , Cigoto/crecimiento & desarrollo
9.
J Invest Dermatol ; 126(9): 2111-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16691193

RESUMEN

To investigate whether the frequency of the BRAF(V600E) (V-raf murine sarcoma virus oncogene homolog B1) mutation in melanocytic nevi is associated with sun exposure patterns, we examined 120 acquired melanocytic nevi excised from various anatomic sites, including glabrous skin, as well as 62 congenital nevi. We used a new mutation detection system based on the shifted termination assay, called Mutector, which was able to detect only 5% of heterozygous mutant cells within the samples. We detected the mutation in 105/120 (87.5%) acquired nevi and 43/62 (69.4%) congenital nevi. Notably, we found the mutation in 35/43 (81.4%) acquired nevi excised from glabrous skin and genitalia. These results strongly suggest that UV light is not necessarily required for the acquisition of the BRAF(V600E) mutation, and suggest that non-mutagenic effects of UV light to melanocytes may be more important in the nevogenesis. Additionally, we showed heterogeneous distribution of BRAF-mutated cells within the lesions of small congenital nevi by a combination of laser microdissection and direct sequencing. Finally, we found low frequency of BRAF(V600E) mutation (6/20, 30.0%) in medium-sized congenital nevi. Most of these nevi with wild-type BRAF had neroblastoma ras viral oncogene homolog mutations (9/14, 64.3%), suggesting different pathogenesis of medium-sized congenital nevi from acquired nevi and small congenital nevi.


Asunto(s)
Nevo/genética , Mutación Puntual , Proteínas Proto-Oncogénicas B-raf/genética , Luz Solar/efectos adversos , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Pruebas Genéticas , Genitales , Humanos , Lactante , Masculino , Melanocitos/patología , Melanocitos/efectos de la radiación , Microdisección , Nevo/congénito , Nevo/patología , Piel/patología
10.
PLoS One ; 8(10): e76478, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24130776

RESUMEN

Zinc finger nuclease (ZFN) is a powerful tool for genome editing. ZFN-encoding plasmid DNA expression systems have been recently employed for the generation of gene knockout (KO) pigs, although one major limitation of this technology is the use of potentially harmful genome-integrating plasmid DNAs. Here we describe a simple, non-integrating strategy for generating KO pigs using ZFN-encoding mRNA. The interleukin-2 receptor gamma (IL2RG) gene was knocked out in porcine fetal fibroblasts using ZFN-encoding mRNAs, and IL2RG KO pigs were subsequently generated using these KO cells through somatic cell nuclear transfer (SCNT). The resulting IL2RG KO pigs completely lacked a thymus and were deficient in T and NK cells, similar to human X-linked SCID patients. Our findings demonstrate that the combination of ZFN-encoding mRNAs and SCNT provides a simple robust method for producing KO pigs without genomic integration.


Asunto(s)
Desoxirribonucleasas/metabolismo , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes/métodos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Dedos de Zinc , Animales , Secuencia de Bases , Separación Celular , Células Cultivadas , Células Clonales , Desoxirribonucleasas/química , Fibroblastos/citología , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Fenotipo , ARN Mensajero/genética , Ratas , Porcinos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda