Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Ther ; 26(5): 1255-1265, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29606506

RESUMEN

X-linked severe combined immunodeficiency (X-SCID) has been successfully treated by hematopoietic stem cell (HSC) transduction with retroviral vectors expressing the interleukin-2 receptor subunit gamma gene (IL2RG), but several patients developed malignancies due to vector integration near cellular oncogenes. This adverse side effect could in principle be avoided by accurate IL2RG gene editing with a vector that does not contain a functional promoter or IL2RG gene. Here, we show that adeno-associated virus (AAV) gene editing vectors can insert a partial Il2rg cDNA at the endogenous Il2rg locus in X-SCID murine bone marrow cells and that these ex vivo-edited cells repopulate transplant recipients and produce CD4+ and CD8+ T cells. Circulating, edited lymphocytes increased over time and appeared in secondary transplant recipients, demonstrating successful editing in long-term repopulating cells. Random vector integration events were nearly undetectable, and malignant transformation of the transplanted cells was not observed. Similar editing frequencies were observed in human hematopoietic cells. Our results demonstrate that therapeutically relevant HSC gene editing can be achieved by AAV vectors in the absence of site-specific nucleases and suggest that this may be a safe and effective therapy for hematopoietic diseases where in vivo selection can increase edited cell numbers.


Asunto(s)
Dependovirus/genética , Edición Génica , Vectores Genéticos/genética , Subunidad gamma Común de Receptores de Interleucina/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Alelos , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Orden Génico , Terapia Genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunoterapia Adoptiva , Ratones , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
2.
Mol Ther ; 21(6): 1232-41, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23629003

RESUMEN

The clinical use of human pluripotent stem cells and their derivatives is limited by the rejection of transplanted cells due to differences in their human leukocyte antigen (HLA) genes. This has led to the proposed use of histocompatible, patient-specific stem cells; however, the preparation of many different stem cell lines for clinical use is a daunting task. Here, we develop two distinct genetic engineering approaches that address this problem. First, we use a combination of gene targeting and mitotic recombination to derive HLA-homozygous embryonic stem cell (ESC) subclones from an HLA-heterozygous parental line. A small bank of HLA-homozygous stem cells with common haplotypes would match a significant proportion of the population. Second, we derive HLA class I-negative cells by targeted disruption of both alleles of the Beta-2 Microglobulin (B2M) gene in ESCs. Mixed leukocyte reactions and peptide-specific HLA-restricted CD8(+) T cell responses were reduced in class I-negative cells that had undergone differentiation in embryoid bodies. These B2M(-/-) ESCs could act as universal donor cells in applications where the transplanted cells do not express HLA class II genes. Both approaches used adeno-associated virus (AAV) vectors for efficient gene targeting in the absence of potentially genotoxic nucleases, and produced pluripotent, transgene-free cell lines.


Asunto(s)
Antígenos HLA/genética , Células Madre Pluripotentes/citología , Alelos , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Línea Celular , Células Cultivadas , Dependovirus/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Marcación de Gen , Ingeniería Genética , Vectores Genéticos , Antígenos HLA/metabolismo , Haplotipos , Histocompatibilidad/genética , Homocigoto , Humanos , Células Madre Pluripotentes/metabolismo , Recombinación Genética , Microglobulina beta-2/genética , Microglobulina beta-2/metabolismo
3.
Mol Ther ; 18(6): 1192-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20407427

RESUMEN

Precise genetic manipulation of human pluripotent stem cells will be required to realize their scientific and therapeutic potential. Here, we show that adeno-associated virus (AAV) gene targeting vectors can be used to genetically engineer human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Different types of sequence-specific changes, including the creation and correction of mutations, were introduced into the human HPRT1 and HMGA1 genes (HPRT1 mutations being responsible for Lesch-Nyhan syndrome). Gene targeting occurred at high frequencies in both ESCs and iPSCs, with over 1% of all colony-forming units (CFUs) undergoing targeting in some experiments. AAV vectors could also be used to target genes in human fibroblasts that were subsequently used to derive iPSCs. Accurate and efficient targeting took place with minimal or no cytotoxicity, and most of the gene-targeted stem cells produced were euploid and pluripotent.


Asunto(s)
Dependovirus/genética , Marcación de Gen , Ingeniería Genética , Células Madre Pluripotentes , Humanos , Hipoxantina Fosforribosiltransferasa/genética
4.
Mol Ther ; 16(1): 187-93, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17955022

RESUMEN

Mesenchymal stem cells (MSCs) are adult cells with the capacity to differentiate into multiple cell types, including bone, fat, cartilage, and muscle cells. In order to effectively utilize autologous MSCs in cell-based therapies, precise genetic manipulations are required to eliminate the effects of disease-causing mutations. We previously used adeno-associated virus (AAV) vectors to target and inactivate mutant COL1A1 genes in MSCs from individuals with the brittle bone disorder, osteogenesis imperfecta (OI). Here we have used AAV vectors to inactivate mutant COL1A2 genes in OI MSCs, thereby demonstrating that both type I collagen genes responsible for OI can be successfully targeted. We incorporated improved vector designs so as to minimize the consequences of random integration, facilitate the removal of potential antigens, and avoid unwanted exon skipping. MSCs targeted at mutant COL1A2 alleles produced normal type I procollagen and formed bone, thereby demonstrating their therapeutic potential.


Asunto(s)
Alelos , Colágeno/antagonistas & inhibidores , Colágeno/genética , Marcación de Gen , Células Madre Mesenquimatosas/metabolismo , Osteogénesis Imperfecta/genética , Osteogénesis Imperfecta/terapia , Mutación Puntual , Adolescente , Sustitución de Aminoácidos/genética , Animales , Línea Celular , Células Cultivadas , Niño , Colágeno/biosíntesis , Colágeno Tipo I , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Osteogénesis Imperfecta/metabolismo
5.
Nat Biotechnol ; 24(8): 1022-6, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16878127

RESUMEN

Therapeutic gene delivery typically involves the addition of a transgene expression cassette to mutant cells. This approach is complicated by transgene silencing, aberrant transcriptional regulation and insertional mutagenesis. An alternative strategy is to correct mutations through homologous recombination, allowing for normal regulation of gene expression from the endogenous locus. Adeno-associated virus (AAV) vectors containing single-stranded DNA efficiently transduce cells in vivo and have been shown to target homologous chromosomal sequences in cultured cells. To determine whether AAV-mediated gene targeting can occur in vivo, we developed a mouse model that contains a mutant, nuclear-localized lacZ gene inserted at the ubiquitously expressed ROSA26 locus. Foci of beta-galactosidase-positive hepatocytes were observed in these mice after injection with an AAV vector containing a lacZ gene fragment, and precise correction of the 4-bp deletion was demonstrated by gene sequencing. We also used AAV gene-targeting vectors to correct the naturally occurring GusB gene mutation responsible for murine mucopolysaccharidosis type VII.


Asunto(s)
Adenoviridae/genética , Marcación de Gen/métodos , Ingeniería Genética/métodos , Vectores Genéticos/genética , Transfección/métodos , Animales , Ratones , Ratones Endogámicos C57BL
7.
Nat Biotechnol ; 35(8): 765-772, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28504668

RESUMEN

Polymorphisms in the human leukocyte antigen (HLA) class I genes can cause the rejection of pluripotent stem cell (PSC)-derived products in allogeneic recipients. Disruption of the Beta-2 Microglobulin (B2M) gene eliminates surface expression of all class I molecules, but leaves the cells vulnerable to lysis by natural killer (NK) cells. Here we show that this 'missing-self' response can be prevented by forced expression of minimally polymorphic HLA-E molecules. We use adeno-associated virus (AAV)-mediated gene editing to knock in HLA-E genes at the B2M locus in human PSCs in a manner that confers inducible, regulated, surface expression of HLA-E single-chain dimers (fused to B2M) or trimers (fused to B2M and a peptide antigen), without surface expression of HLA-A, B or C. These HLA-engineered PSCs and their differentiated derivatives are not recognized as allogeneic by CD8+ T cells, do not bind anti-HLA antibodies and are resistant to NK-mediated lysis. Our approach provides a potential source of universal donor cells for applications where the differentiated derivatives lack HLA class II expression.


Asunto(s)
Antígenos HLA/inmunología , Células Asesinas Naturales/inmunología , Células Madre Pluripotentes/inmunología , Trasplantes/inmunología , Animales , Femenino , Rechazo de Injerto/inmunología , Antígenos HLA/química , Antígenos HLA/genética , Humanos , Ratones , Células Madre Pluripotentes/química , Células Madre Pluripotentes/citología , Trasplantes/química , Trasplantes/citología
8.
Hum Gene Ther ; 16(4): 522-6, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15871683

RESUMEN

Adeno-associated virus (AAV) vectors can be used to introduce site-specific mutations into homologous chromosomal sequences. There are many potential applications of this technique, but the process of AAV-mediated gene targeting and factors that influence targeting efficiency are not completely understood. We investigated the dependence of AAV-mediated gene targeting on the host cell-cycle status. The frequency of gene targeting by AAV vectors was compared in dividing and serum-arrested normal human fibroblast cultures. Gene targeting occurred in arrested fibroblast cultures at 0.15 to 1.1% the frequency of dividing cultures, and only took place in cells that had undergone DNA synthesis. Gene targeting was also reduced when DNA synthesis was inhibited by hydroxyurea.


Asunto(s)
Ciclo Celular/genética , Dependovirus/genética , Marcación de Gen/métodos , Vectores Genéticos/genética , Bromodesoxiuridina/metabolismo , Células Cultivadas , Medio de Cultivo Libre de Suero , ADN/biosíntesis , Replicación del ADN , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Hidroxiurea/farmacología , Hipoxantina Fosforribosiltransferasa/genética , Masculino
9.
Cloning Stem Cells ; 6(1): 31-6, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15107244

RESUMEN

Gene-targeted livestock can be created by combining ex vivo manipulation of cultured nuclear donor cells with cloning by nuclear transfer. However, this process can be limited by the low gene targeting frequencies obtained by transfection methods, and the limited ex vivo life span of the normal nuclear donor cells. We have developed an alternative gene targeting method based on the delivery of linear, single-stranded DNA molecules by adeno-associated virus (AAV) vectors, which can be used to introduce a variety of different mutations at single copy loci in normal human cells. Here we show that AAV vectors can efficiently target the PRNP gene encoding the prion protein PrP in bovine fetal fibroblasts, which can be used as nuclear donors to clone cattle. Cattle with both PRNP genes disrupted should be resistant to bovine spongiform encephalopathy.


Asunto(s)
Marcación de Gen/métodos , Proteínas del Tejido Nervioso/genética , Priones/genética , Animales , Bovinos , Línea Celular , Clonación de Organismos , Dependovirus/genética , Encefalopatía Espongiforme Bovina/prevención & control , Vectores Genéticos , Humanos , Técnicas de Transferencia Nuclear
11.
Cell Stem Cell ; 11(5): 615-9, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-23084023

RESUMEN

Human trisomies can alter cellular phenotypes and produce congenital abnormalities such as Down syndrome (DS). Here we have generated induced pluripotent stem cells (iPSCs) from DS fibroblasts and introduced a TKNEO transgene into one copy of chromosome 21 by gene targeting. When selecting against TKNEO, spontaneous chromosome loss was the most common cause for survival, with a frequency of ~10(-4), while point mutations, epigenetic silencing, and TKNEO deletions occurred at lower frequencies in this unbiased comparison of inactivating mutations. Mitotic recombination events resulting in extended loss of heterozygosity were not observed in DS iPSCs. The derived, disomic cells proliferated faster and produced more endothelia in vivo than their otherwise isogenic trisomic counterparts, but in vitro hematopoietic differentiation was not consistently altered. Our study describes a targeted removal of a human trisomy, which could prove useful in both clinical and research applications.


Asunto(s)
Síndrome de Down/genética , Células Madre Pluripotentes Inducidas/citología , Trisomía , Diferenciación Celular , Cromosomas Humanos Par 21 , Epigénesis Genética , Fibroblastos/metabolismo , Marcación de Gen , Humanos , Células Madre Pluripotentes Inducidas/metabolismo
12.
Nat Protoc ; 6(4): 482-501, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21455185

RESUMEN

Gene targeting with adeno-associated virus (AAV) vectors has been demonstrated in multiple human cell types, with targeting frequencies ranging from 10(-5) to 10(-2) per infected cell. These targeting frequencies are 1-4 logs higher than those obtained by conventional transfection or electroporation approaches. A wide variety of different types of mutations can be introduced into chromosomal loci with high fidelity and without genotoxicity. Here we provide a detailed protocol for gene targeting in human cells with AAV vectors. We describe methods for vector design, stock preparation and titration. Optimized transduction protocols are provided for human pluripotent stem cells, mesenchymal stem cells, fibroblasts and transformed cell lines, as well as a method for identifying targeted clones by Southern blots. This protocol (from vector design through a single round of targeting and screening) can be completed in ∼10 weeks; each subsequent round of targeting and screening should take an additional 7 weeks.


Asunto(s)
Dependovirus/genética , Marcación de Gen/métodos , Animales , Células Cultivadas , Vectores Genéticos , Humanos , Ratones , Transducción Genética/métodos
13.
PLoS One ; 5(1): e8763, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20098701

RESUMEN

BACKGROUND: Inherited and acquired retinal degenerations are frequent causes of visual impairment and photoreceptor cell replacement therapy may restore visual function to these individuals. To provide a source of new retinal neurons for cell based therapies, we developed methods to derive retinal progenitors from human ES cells. METHODOLOGY/PHYSICAL FINDINGS: In this report we have used a similar method to direct induced pluripotent stem cells (iPS) from human fibroblasts to a retinal progenitor fate, competent to generate photoreceptors. We also found we could purify the photoreceptors derived from the iPS cells using fluorescence activated cell sorting (FACS) after labeling photoreceptors with a lentivirus driving GFP from the IRBP cis-regulatory sequences. Moreover, we found that when we transplanted the FACS purified iPSC derived photoreceptors, they were able to integrate into a normal mouse retina and express photoreceptor markers. CONCLUSIONS: This report provides evidence that enriched populations of human photoreceptors can be derived from iPS cells.


Asunto(s)
Células Fotorreceptoras de Vertebrados/metabolismo , Células Madre Pluripotentes/metabolismo , Trasplante de Células Madre , Citometría de Flujo , Humanos , Inmunohistoquímica , Reacción en Cadena de la Polimerasa
14.
Hum Gene Ther ; 19(9): 907-14, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18680404

RESUMEN

Gene targeting is a powerful technique for manipulating the human genome, but few studies have directly compared the targeting frequencies of various types of vector constructs. Here we show that similar targeting constructs are able to insert nucleotides at the homologous chromosomal target locus more efficiently than they can delete nucleotides, and combination insertion/deletion vectors appear to target at intermediate frequencies. This holds true for deletions ranging from 1 to 334 bp and insertions ranging from 1 to 1332 bp. In addition, vectors designed to inactivate the human hypoxanthine phosphoribosyltransferase gene (HPRT) by deleting nucleotides often produced rearrangements at the target locus that in many cases were due to insertions of multimerized vector constructs, effectively converting a deletion vector into an insertion vector. These findings were obtained when adeno-associated virus vectors were used to efficiently deliver single-stranded DNA targeting constructs, but the same phenomenon was also observed when transfecting linearized double-stranded plasmids. Thus human cells distinguish between deletion and insertion vectors and process their recombination intermediates differently, presumably at the heteroduplex stage, with implications for the design of gene-targeting vectors and the evolution of human genomes.


Asunto(s)
Eliminación de Gen , Marcación de Gen/métodos , Mutagénesis Insercional , Fosfatasa Alcalina/genética , Secuencia de Bases , Línea Celular , Dependovirus/genética , Vectores Genéticos , Genoma Humano , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Mutación , Plásmidos/genética , Transfección
15.
J Cell Biol ; 181(6): 913-20, 2008 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-18559665

RESUMEN

The SCF(FBW7) ubiquitin ligase degrades proteins involved in cell division, growth, and differentiation and is commonly mutated in cancers. The Fbw7 locus encodes three protein isoforms that occupy distinct subcellular localizations, suggesting that each has unique functions. We used gene targeting to create isoform-specific Fbw7-null mutations in human cells and found that the nucleoplasmic Fbw7alpha isoform accounts for almost all Fbw7 activity toward cyclin E, c-Myc, and sterol regulatory element binding protein 1. Cyclin E sensitivity to Fbw7 varies during the cell cycle, and this correlates with changes in cyclin E-cyclin-dependent kinase 2 (CDK2)-specific activity, cyclin E autophosphorylation, and CDK2 inhibitory phosphorylation. These data suggest that oscillations in cyclin E-CDK2-specific activity during the cell cycle regulate the timing of cyclin E degradation. Moreover, they highlight the utility of adeno-associated virus-mediated gene targeting in functional analyses of complex loci.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Proteínas F-Box/metabolismo , Procesamiento Proteico-Postraduccional , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular Tumoral , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Estabilidad de Enzimas , Proteína 7 que Contiene Repeticiones F-Box-WD , Marcación de Gen , Humanos , Isoenzimas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Especificidad por Sustrato
16.
Mol Ther ; 15(10): 1827-33, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17622243

RESUMEN

Human embryonic stem cells (hESCs) are important tools for the study of stem cell biology and may ultimately be used in cellular therapies and regenerative medicine. For hESCs to achieve their potential, stable genetic modification of the hESC genome will be required. Here we have studied the transduction of hESCs by vectors based on foamy virus (FV), an integrating retrovirus with no known pathogenicity. We find that hESCs and also ESCs derived from rhesus monkeys can be efficiently transduced by FV vectors at frequencies of 14-48%. Integration of FV vector DNA was demonstrated by Southern blot analysis, and stable expression was observed from a single integrated provirus in several clones. Transduced hESCs expressed markers characteristic of undifferentiated cells, differentiated and expressed markers from all three germ layers after serum exposure, and formed teratomas with persistent transgene expression in differentiated cells. Thus, FV vectors are promising tools for the genetic modification of hESCs.


Asunto(s)
Células Madre Embrionarias/virología , Vectores Genéticos , Spumavirus/genética , Transducción Genética , Secuencia de Bases , Southern Blotting , Línea Celular , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Técnicas de Transferencia de Gen , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
J Virol ; 77(24): 13136-45, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14645570

RESUMEN

The molecular mechanisms responsible for random integration and gene targeting by recombinant adeno-associated virus (AAV) vectors are largely unknown, and whether vectors derived from autonomous parvoviruses transduce cells by similar pathways has not been investigated. In this report, we constructed vectors based on the autonomous parvovirus minute virus of mice (MVM) that were designed to introduce a neomycin resistance expression cassette (neo) into the X-linked human hypoxanthine phosphoribosyl transferase (HPRT) locus. High-titer, replication-incompetent MVM vector stocks were generated with a two-plasmid transfection system that preserved the wild-type characteristic of packaging only one DNA strand. Vectors with inserts in the forward or reverse orientations packaged noncoding or coding strands, respectively. In human HT-1080 cells, MVM vector random integration frequencies (neo(+) colonies) were comparable to those obtained with AAV vectors, and no difference was observed for noncoding and coding strands. HPRT gene-targeting frequencies (HPRT mutant colonies) were lower with MVM vectors, and the noncoding strand frequency was threefold greater than that of the coding strand. Random integration and gene-targeting events were confirmed by Southern blot analysis of G418- and 6-thioguanine (6TG)-resistant clones. In separate experiments, correction of an alkaline phosphatase (AP) gene by gene targeting was nine times more effective with a coding strand vector. The data suggest that single-stranded parvoviral vector genomes are substrates for gene targeting and possibly for random integration as well.


Asunto(s)
Cromosomas Humanos/virología , Marcación de Gen , Vectores Genéticos/genética , Virus Diminuto del Ratón/genética , Integración Viral , Animales , Línea Celular , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Ratones , Virus Diminuto del Ratón/fisiología , Plásmidos , Transducción Genética , Transfección , Replicación Viral
18.
Science ; 303(5661): 1198-201, 2004 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-14976317

RESUMEN

Adult stem cells offer the potential to treat many diseases through a combination of ex vivo genetic manipulation and autologous transplantation. Mesenchymal stem cells (MSCs, also referred to as marrow stromal cells) are adult stem cells that can be isolated as proliferating, adherent cells from bones. MSCs can differentiate into multiple cell types present in several tissues, including bone, fat, cartilage, and muscle, making them ideal candidates for a variety of cell-based therapies. Here, we have used adeno-associated virus vectors to disrupt dominant-negative mutant COL1A1 collagen genes in MSCs from individuals with the brittle bone disorder osteogenesis imperfecta, demonstrating successful gene targeting in adult human stem cells.


Asunto(s)
Colágeno Tipo I/genética , Marcación de Gen , Células Madre Mesenquimatosas/fisiología , Osteogénesis Imperfecta/genética , Osteogénesis Imperfecta/terapia , Alelos , Animales , Células de la Médula Ósea/fisiología , Diferenciación Celular , Células Cultivadas , Colágeno Tipo I/química , Colágeno Tipo I/metabolismo , Dependovirus/genética , Terapia Genética , Vectores Genéticos , Humanos , Kanamicina Quinasa/genética , Masculino , Ratones , Osteogénesis , Mutación Puntual , Recombinación Genética , Trasplante de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA