Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
PLoS Genet ; 11(5): e1005239, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26000857

RESUMO

In vitro disease modeling based on induced pluripotent stem cells (iPSCs) provides a powerful system to study cellular pathophysiology, especially in combination with targeted genome editing and protocols to differentiate iPSCs into affected cell types. In this study, we established zinc-finger nuclease-mediated genome editing in primary fibroblasts and iPSCs generated from a mouse model for radiosensitive severe combined immunodeficiency (RS-SCID), a rare disorder characterized by cellular sensitivity to radiation and the absence of lymphocytes due to impaired DNA-dependent protein kinase (DNA-PK) activity. Our results demonstrate that gene editing in RS-SCID fibroblasts rescued DNA-PK dependent signaling to overcome radiosensitivity. Furthermore, in vitro T-cell differentiation from iPSCs was employed to model the stage-specific T-cell maturation block induced by the disease causing mutation. Genetic correction of the RS-SCID iPSCs restored T-lymphocyte maturation, polyclonal V(D)J recombination of the T-cell receptor followed by successful beta-selection. In conclusion, we provide proof that iPSC-based in vitro T-cell differentiation is a valuable paradigm for SCID disease modeling, which can be utilized to investigate disorders of T-cell development and to validate gene therapy strategies for T-cell deficiencies. Moreover, this study emphasizes the significance of designer nucleases as a tool for generating isogenic disease models and their future role in producing autologous, genetically corrected transplants for various clinical applications.


Assuntos
Diferenciação Celular , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Animais , Proteína Quinase Ativada por DNA/deficiência , Proteína Quinase Ativada por DNA/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Fibroblastos/citologia , Fibroblastos/metabolismo , Genoma , Técnicas de Genotipagem , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Camundongos , Células NIH 3T3 , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Fenótipo , Proteínas Quinases/genética , Linfócitos T/metabolismo , Dedos de Zinco
2.
Methods ; 53(4): 339-46, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21185378

RESUMO

Zinc-finger nucleases (ZFNs) are designer nucleases capable of cleaving a prespecified target DNA within complex genomes. ZFNs consist of a non-specific endonuclease domain fused to an engineered DNA-binding domain that tethers the nuclease activity to the chosen chromosomal site. The endonuclease-induced DNA double strand break triggers a cellular DNA damage response, resulting in double strand break repair by either accurate homologous recombination (HR) or error-prone non-homologous end-joining (NHEJ). Thus, ZFNs are powerful tools for targeted genome engineering in a variety of mammalian cell types, including embryonic (ESCs) and induced pluripotent stem cells (iPSCs). As a paradigm for genome editing in pluripotent stem cells, we describe the use of ZFNs in murine ESCs for generating knockout alleles by NHEJ without selection or by HR employing different selection schemes.


Assuntos
Desoxirribonucleases/genética , Células-Tronco Embrionárias/fisiologia , Técnicas de Introdução de Genes , Técnicas de Silenciamento de Genes , Genoma , Dedos de Zinco/genética , Animais , Técnicas de Cultura de Células , Células Cultivadas , Meios de Cultura , Reparo do DNA , Desoxirribonucleases/metabolismo , Células-Tronco Embrionárias/citologia , Ensaios Enzimáticos , Deleção de Genes , Engenharia Genética/métodos , Genótipo , Camundongos , Transfecção/métodos
4.
Nat Biotechnol ; 33(8): 842-4, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26192319

RESUMO

Production of glycoprotein therapeutics in Chinese hamster ovary (CHO) cells is limited by the cells' generic capacity for N-glycosylation, and production of glycoproteins with desirable homogeneous glycoforms remains a challenge. We conducted a comprehensive knockout screen of glycosyltransferase genes controlling N-glycosylation in CHO cells and constructed a design matrix that facilitates the generation of desired glycosylation, such as human-like α2,6-linked sialic acid capping. This engineering approach will aid the production of glycoproteins with improved properties and therapeutic potential.


Assuntos
Glicoproteínas , Engenharia de Proteínas/métodos , Proteínas Recombinantes , Animais , Células CHO , Cricetinae , Cricetulus , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
Hum Gene Ther ; 24(1): 67-77, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23072634

RESUMO

Parameters that regulate or affect the cell cycle or the DNA repair choice between non-homologous end-joining and homology-directed repair (HDR) are excellent targets to enhance therapeutic gene targeting. Here, we have evaluated the impact of five cell-cycle modulating drugs on targeted genome engineering mediated by DNA double-strand break (DSB)-inducing nucleases, such as zinc-finger nucleases (ZFNs). For a side-by-side comparison, we have established four reporter cell lines by integrating a mutated EGFP gene into either three transformed human cell lines or primary umbilical cord-derived mesenchymal stromal cells (UC-MSCs). After treatment with different cytostatic drugs, cells were transduced with adeno-associated virus (AAV) vectors that encode a nuclease or a repair donor to rescue EGFP expression through DSB-induced HDR. We show that transient cell-cycle arrest increased AAV transduction and AAV-mediated HDR up to six-fold in human cell lines and ten-fold in UC-MSCs, respectively. Targeted gene correction was observed in up to 34% of transduced cells. Both the absolute and the relative gene-targeting frequencies were dependent on the cell type, the cytostatic drug, the vector dose, and the nuclease. Treatment of cells with the cyclin-dependent kinase inhibitor indirubin-3'-monoxime was especially promising as this compound combined high stimulatory effects with minimal cytotoxicity. In conclusion, indirubin-3'-monoxime significantly improved AAV transduction and the efficiency of AAV/ZFN-mediated gene targeting and may thus represent a promising compound to enhance DSB-mediated genome engineering in human stem cells, such as UC-MSCs, which hold great promise for future clinical applications.


Assuntos
Dependovirus/genética , Marcação de Genes/métodos , Engenharia Genética/métodos , Vetores Genéticos/genética , Indóis/uso terapêutico , Oximas/uso terapêutico , Transdução Genética/métodos , Western Blotting , Pontos de Checagem do Ciclo Celular/fisiologia , Linhagem Celular , Primers do DNA/genética , Reparo do DNA/fisiologia , Desoxirribonucleases/metabolismo , Genótipo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células-Tronco Mesenquimais , Reação em Cadeia da Polimerase em Tempo Real
6.
Hum Gene Ther ; 22(8): 925-33, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21631241

RESUMO

Zinc-finger nucleases (ZFNs) are a powerful tool that can be used to edit the human genome ad libitum. The technology has experienced remarkable development in the last few years with regard to both the target site specificity and the engineering platforms used to generate zinc-finger proteins. As a result, two phase I clinical trials aimed at knocking out the CCR5 receptor in T cells isolated from HIV patients to protect these lymphocytes from infection with the virus have been initiated. Moreover, ZFNs have been successfully employed to knockout or correct disease-related genes in human stem cells, including hematopoietic precursor cells and induced pluripotent stem cells. Targeted genome engineering approaches in multipotent and pluripotent stem cells hold great promise for future strategies geared toward correcting inborn mutations for personalized cell replacement therapies. This review describes how ZFNs have been applied to models of gene therapy, discusses the opportunities and the risks associated with this novel technology, and suggests future directions for their safe application in therapeutic genome engineering.


Assuntos
Endonucleases/genética , Terapia Genética/tendências , Dedos de Zinco , Engenharia Genética/métodos , Terapia Genética/métodos , Genoma Humano , Infecções por HIV/terapia , Humanos , Receptores CCR5/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA