Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Añadir filtros








Intervalo de año
1.
Protein & Cell ; (12): 601-611, 2017.
Artículo en Inglés | WPRIM | ID: wpr-756965

RESUMEN

Targeted point mutagenesis through homologous recombination has been widely used in genetic studies and holds considerable promise for repairing disease-causing mutations in patients. However, problems such as mosaicism and low mutagenesis efficiency continue to pose challenges to clinical application of such approaches. Recently, a base editor (BE) system built on cytidine (C) deaminase and CRISPR/Cas9 technology was developed as an alternative method for targeted point mutagenesis in plant, yeast, and human cells. Base editors convert C in the deamination window to thymidine (T) efficiently, however, it remains unclear whether targeted base editing in mouse embryos is feasible. In this report, we generated a modified high-fidelity version of base editor 2 (HF2-BE2), and investigated its base editing efficacy in mouse embryos. We found that HF2-BE2 could convert C to T efficiently, with up to 100% biallelic mutation efficiency in mouse embryos. Unlike BE3, HF2-BE2 could convert C to T on both the target and non-target strand, expanding the editing scope of base editors. Surprisingly, we found HF2-BE2 could also deaminate C that was proximal to the gRNA-binding region. Taken together, our work demonstrates the feasibility of generating point mutations in mouse by base editing, and underscores the need to carefully optimize base editing systems in order to eliminate proximal-site deamination.


Asunto(s)
Animales , Humanos , Ratones , Desaminasas APOBEC-1 , Genética , Metabolismo , Proteínas Bacterianas , Genética , Metabolismo , Secuencia de Bases , Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Citidina , Genética , Metabolismo , Transferencia de Embrión , Embrión de Mamíferos , Endonucleasas , Genética , Metabolismo , Edición Génica , Métodos , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones Endogámicos C57BL , Microinyecciones , Plásmidos , Química , Metabolismo , Mutación Puntual , Genética , Metabolismo , Timidina , Genética , Metabolismo , Cigoto , Metabolismo , Trasplante
2.
Protein & Cell ; (12): 811-822, 2017.
Artículo en Inglés | WPRIM | ID: wpr-756922

RESUMEN

β-Thalassemia is a global health issue, caused by mutations in the HBB gene. Among these mutations, HBB -28 (A>G) mutations is one of the three most common mutations in China and Southeast Asia patients with β-thalassemia. Correcting this mutation in human embryos may prevent the disease being passed onto future generations and cure anemia. Here we report the first study using base editor (BE) system to correct disease mutant in human embryos. Firstly, we produced a 293T cell line with an exogenous HBB -28 (A>G) mutant fragment for gRNAs and targeting efficiency evaluation. Then we collected primary skin fibroblast cells from a β-thalassemia patient with HBB -28 (A>G) homozygous mutation. Data showed that base editor could precisely correct HBB -28 (A>G) mutation in the patient's primary cells. To model homozygous mutation disease embryos, we constructed nuclear transfer embryos by fusing the lymphocyte or skin fibroblast cells with enucleated in vitro matured (IVM) oocytes. Notably, the gene correction efficiency was over 23.0% in these embryos by base editor. Although these embryos were still mosaic, the percentage of repaired blastomeres was over 20.0%. In addition, we found that base editor variants, with narrowed deamination window, could promote G-to-A conversion at HBB -28 site precisely in human embryos. Collectively, this study demonstrated the feasibility of curing genetic disease in human somatic cells and embryos by base editor system.


Asunto(s)
Femenino , Humanos , Desaminasas APOBEC-1 , Genética , Metabolismo , Secuencia de Bases , Blastómeros , Biología Celular , Metabolismo , Sistemas CRISPR-Cas , Embrión de Mamíferos , Metabolismo , Patología , Fibroblastos , Metabolismo , Patología , Edición Génica , Métodos , Expresión Génica , Células HEK293 , Heterocigoto , Homocigoto , Mutación Puntual , Cultivo Primario de Células , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Globinas beta , Genética , Metabolismo , Talasemia beta , Genética , Metabolismo , Patología , Terapéutica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA