Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cells Transl Med ; 2(12): 935-45, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24167318

RESUMEN

Integrative gene transfer using retroviruses to express reprogramming factors displays high efficiency in generating induced pluripotent stem cells (iPSCs), but the value of the method is limited because of the concern over mutagenesis associated with random insertion of transgenes. Site-specific integration into a preselected locus by engineered zinc-finger nuclease (ZFN) technology provides a potential way to overcome the problem. Here, we report the successful reprogramming of human fibroblasts into a state of pluripotency by baculoviral transduction-mediated, site-specific integration of OKSM (Oct3/4, Klf4, Sox2, and c-myc) transcription factor genes into the AAVS1 locus in human chromosome 19. Two nonintegrative baculoviral vectors were used for cotransduction, one expressing ZFNs and another as a donor vector encoding the four transcription factors. iPSC colonies were obtained at a high efficiency of 12% (the mean value of eight individual experiments). All characterized iPSC clones carried the transgenic cassette only at the ZFN-specified AAVS1 locus. We further demonstrated that when the donor cassette was flanked by heterospecific loxP sequences, the reprogramming genes in iPSCs could be replaced by another transgene using a baculoviral vector-based Cre recombinase-mediated cassette exchange system, thereby producing iPSCs free of exogenous reprogramming factors. Although the use of nonintegrating methods to generate iPSCs is rapidly becoming a standard approach, methods based on site-specific integration of reprogramming factor genes as reported here hold the potential for efficient generation of genetically amenable iPSCs suitable for future gene therapy applications.


Asunto(s)
Baculoviridae/genética , Reprogramación Celular , Endonucleasas/metabolismo , Fibroblastos/metabolismo , Vectores Genéticos , Células Madre Pluripotentes Inducidas/metabolismo , Factores de Transcripción/metabolismo , Transducción Genética , Dedos de Zinc , Diferenciación Celular , Línea Celular , Cromosomas Humanos Par 19 , Endonucleasas/genética , Regulación del Desarrollo de la Expresión Génica , Genotipo , Humanos , Integrasas/genética , Integrasas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/genética , Transfección
2.
J Gene Med ; 15(10): 384-95, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24105820

RESUMEN

BACKGROUND: The AAVS1 locus is viewed as a 'safe harbor' for transgene insertion into human genome. In the present study, we report a new method for AAVS1 targeting in human-induced pluripotent stem cells (hiPSCs). METHODS: We have developed two baculoviral transduction systems: one to deliver zinc finger nuclease (ZFN) and a DNA donor template for site-specific gene insertion and another to mediate Cre recombinase-mediated cassette exchange system to replace the inserted transgene with a new transgene. RESULTS: Our ZFN system provided the targeted integration efficiency of a Neo-EGFP cassette of 93.8% in G418-selected, stable hiPSC colonies. Southern blotting analysis of 20 AASV1 targeted colonies revealed no random integration events. Among 24 colonies examined for mono- or biallelic AASV1 targeting, 25% of them were biallelically modified. The selected hiPSCs displayed persistent enhanced green fluorescent protein expression and continued the expression of stem cell pluripotency markers. The hiPSCs maintained the ability to differentiate into three germ lineages in derived embryoid bodies and transgene expression was retained in the differentiated cells. After pre-including the loxP-docking sites into the Neo-EGFP cassette, we demonstrated that a baculovirus-Cre/loxP system could be used to facilitate the replacement of the Neo-EGFP cassette with another transgene cassette at the AAVS1 locus. CONCLUSIONS: Given high targeting efficiency, stability in expression of inserted transgene and flexibility in transgene exchange, the approach reported in the present study holds potential for generating genetically-modified human pluripotent stem cells suitable for developmental biology research, drug development, regenerative medicine and gene therapy.


Asunto(s)
Baculoviridae/genética , Endonucleasas/genética , Expresión Génica , Vectores Genéticos/genética , Células Madre Pluripotentes Inducidas/metabolismo , Transgenes , Dedos de Zinc/genética , Endonucleasas/metabolismo , Orden Génico , Genes Reporteros , Sitios Genéticos , Recombinación Homóloga , Humanos , Mutagénesis Insercional
3.
Nucleic Acids Res ; 41(19): e180, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23945944

RESUMEN

Safety and reliability of transgene integration in human genome continue to pose challenges for stem cell-based gene therapy. Here, we report a baculovirus-transcription activator-like effector nuclease system for AAVS1 locus-directed homologous recombination in human induced pluripotent stem cells (iPSCs). This viral system, when optimized in human U87 cells, provided a targeted integration efficiency of 95.21% in incorporating a Neo-eGFP cassette and was able to mediate integration of DNA insert up to 13.5 kb. In iPSCs, targeted integration with persistent transgene expression was achieved without compromising genomic stability. The modified iPSCs continued to express stem cell pluripotency markers and maintained the ability to differentiate into three germ lineages in derived embryoid bodies. Using a baculovirus-Cre/LoxP system in the iPSCs, the Neo-eGFP cassette at the AAVS1 locus could be replaced by a Hygro-mCherry cassette, demonstrating the feasibility of cassette exchange. Moreover, as assessed by measuring γ-H2AX expression levels, genome toxicity associated with chromosomal double-strand breaks was not detectable after transduction with moderate doses of baculoviral vectors expressing transcription activator-like effector nucleases. Given high targeted integration efficiency, flexibility in transgene exchange and low genome toxicity, our baculoviral transduction-based approach offers great potential and attractive option for precise genetic manipulation in human pluripotent stem cells.


Asunto(s)
Baculoviridae/genética , Proteínas de Unión al ADN/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Marcación de Gen/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Transducción Genética , Transgenes , Línea Celular Tumoral , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/genética , Desoxirribonucleasas de Localización Especificada Tipo II/genética , Vectores Genéticos , Células HEK293 , Humanos , Integrasas/metabolismo , Mutación , Proteínas Recombinantes de Fusión/metabolismo
4.
Mol Ther ; 21(8): 1621-30, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23752308

RESUMEN

Intravenously injected neural stem cells (NSCs) can infiltrate both primary and metastatic tumor sites; thus, they are attractive tumor-targeting vehicles for delivering anticancer agents. However, because the systemic distribution of the injected NSCs involves normal organs and might induce off-target actions leading to unintended side effects, clinical applications of this approach is impeded. Given that the vesicular stomatitis virus glycoprotein (VSV-G) can promote the formation of multinucleated syncytia to kill cells in a pH-dependent manner, we engineered a pH sensor of VSV-G and generated a novel VSV-G mutant that efficiently promotes syncytium formation at the tumor extracellular pH (pHe) but not at pH 7.4. Using transduced NSCs derived from induced pluripotent stem cells (iPSCs), the VSV-G mutant was delivered into mice with metastatic breast cancers in the lung through tail vein injection. Compared with the conventional stem cell-based gene therapy that uses the herpes simplex virus thymidine kinase (HSVtk) suicide gene, this treatment did not display toxicity to normal non-targeted organs while retaining therapeutic effects in tumor-bearing organs. Our findings demonstrate the effectiveness of a new approach for achieving tumor-selective killing effects following systemic stem cell administration. Its potential in stem cell-based gene therapy for metastatic cancer is worthy of further exploration.


Asunto(s)
Glicoproteínas de Membrana/genética , Neoplasias/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante , Proteínas del Envoltorio Viral/genética , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Muerte Celular , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos , Modelos Animales de Enfermedad , Femenino , Genes Transgénicos Suicidas , Terapia Genética , Células Gigantes/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Células Madre Pluripotentes Inducidas/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Metástasis de la Neoplasia , Neoplasias/mortalidad , Neoplasias/patología , Neoplasias/terapia , Proteínas del Envoltorio Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...