Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nat Methods ; 11(10): 1051-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25152084

RESUMO

Engineered sequence-specific nucleases and donor DNA templates can be customized to edit mammalian genomes via the homologous recombination (HR) pathway. Here we report that the nature of the donor DNA greatly affects the specificity and accuracy of the editing process following site-specific genomic cleavage by transcription activator-like effector nucleases (TALENs) and clustered, regularly interspaced, short palindromic repeats (CRISPR)-Cas9 nucleases. By applying these designer nucleases together with donor DNA delivered as protein-capped adenoviral vector (AdV), free-ended integrase-defective lentiviral vector or nonviral vector templates, we found that the vast majority of AdV-modified human cells underwent scarless homology-directed genome editing. In contrast, a significant proportion of cells exposed to free-ended or to covalently closed HR substrates were subjected to random and illegitimate recombination events. These findings are particularly relevant for genome engineering approaches aiming at high-fidelity genetic modification of human cells.


Assuntos
Adenoviridae/genética , DNA Viral/genética , DNA/química , Desoxirribonucleases/química , Engenharia Genética/métodos , Linhagem Celular , Separação Celular , Marcação de Genes/métodos , Genoma , Células HEK293 , Células HeLa , Humanos , Reação em Cadeia da Polimerase , Recombinação Genética , Sequências Repetitivas de Ácido Nucleico , Reprodutibilidade dos Testes
2.
Methods ; 69(2): 179-87, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24561826

RESUMO

Transcription activator-like effector nucleases (TALENs) are designed to cut the genomic DNA at specific chromosomal positions. The resulting DNA double strand break activates cellular repair pathways that can be harnessed for targeted genome modifications. TALENs thus constitute a powerful tool to interrogate the function of DNA sequences within complex genomes. Moreover, their high DNA cleavage activity combined with a low cytotoxicity make them excellent candidates for applications in human gene therapy. Full exploitation of these large and repeat-bearing nucleases in human cell types will benefit largely from using the adenoviral vector (AdV) technology. The genetic stability and the episomal nature of AdV genomes in conjunction with the availability of a large number of AdV serotypes able to transduce various human cell types make it possible to achieve high-level and transient expression of TALENs in numerous target cells, regardless of their mitotic state. Here, we describe a set of protocols detailing the rescue, propagation and purification of TALEN-encoding AdVs. Moreover, we describe procedures for the characterization and quantification of recombinant viral DNA present in the resulting AdV preparations. The protocols are preceded by information about their underlying principles and applied in the context of second-generation capsid-modified AdVs expressing TALENs targeted to the AAVS1 "safe harbor" locus on human chromosome 19.


Assuntos
Adenoviridae/genética , DNA Viral/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , DNA Viral/administração & dosagem , Endonucleases/genética , Vetores Genéticos/administração & dosagem , Células HEK293 , Células HeLa , Humanos
3.
Nucleic Acids Res ; 41(5): e63, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23275534

RESUMO

The array of genome editing strategies based on targeted double-stranded DNA break formation have recently been enriched through the introduction of transcription activator-like type III effector (TALE) nucleases (TALENs). To advance the testing of TALE-based approaches, it will be crucial to deliver these custom-designed proteins not only into transformed cell types but also into more relevant, chromosomally stable, primary cells. Viral vectors are among the most effective gene transfer vehicles. Here, we investigated the capacity of human immunodeficiency virus type 1- and adenovirus-based vectors to package and deliver functional TALEN genes into various human cell types. To this end, we attempted to assemble particles of these two vector classes, each encoding a monomer of a TALEN pair targeted to a bipartite sequence within the AAVS1 'safe harbor' locus. Vector DNA analyses revealed that adenoviral vectors transferred intact TALEN genes, whereas lentiviral vectors failed to do so, as shown by their heterogeneously sized proviruses in target cells. Importantly, adenoviral vector-mediated TALEN gene delivery resulted in site-specific double-stranded DNA break formation at the intended AAVS1 target site at similarly high levels in both transformed and non-transformed cells. In conclusion, we demonstrate that adenoviral, but not lentiviral, vectors constitute a valuable TALEN gene delivery platform.


Assuntos
Adenoviridae/genética , Proteínas de Bactérias/genética , Desoxirribonucleases/genética , HIV-1/genética , Cromossomos Humanos , Quebras de DNA de Cadeia Dupla , Loci Gênicos , Vetores Genéticos , Células HEK293 , Células HeLa , Humanos , Transdução Genética
4.
Nucleic Acids Res ; 40(5): 1984-99, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22080552

RESUMO

DNA repeats constitute potential sites for the nucleation of secondary structures such as hairpins and cruciforms. Studies performed mostly in bacteria and yeast showed that these noncanonical DNA structures are breakage-prone, making them candidate targets for cellular DNA repair pathways. Possible culprits for fragility at repetitive DNA sequences include replication and transcription as well as the action of structure-specific nucleases. Despite their patent biological relevance, the parameters governing DNA repeat-associated chromosomal transactions remain ill-defined. Here, we established an episomal recombination system based on donor and acceptor complementary DNA templates to investigate the role of direct and inverted DNA repeats in homologous recombination (HR) in mammalian cells. This system allowed us also to ascertain in a stringent manner the impact of repetitive sequence replication on homology-directed gene repair. We found that nonspaced DNA repeats can, per se, engage the HR pathway of the cell and that this process is primarily dependent on their spacing and relative arrangement (i.e. parallel or antiparallel) rather than on their sequence. Indeed, our data demonstrate that contrary to direct and spaced inverted repeats, nonspaced inverted repeats are intrinsically recombinogenic motifs in mammalian cells lending experimental support to their role in genome dynamics in higher eukaryotes.


Assuntos
DNA/química , Sequências Repetidas Invertidas , Reparo de DNA por Recombinação , Animais , Sequência de Bases , Células COS , Linhagem Celular , Chlorocebus aethiops , DNA Cruciforme/química , Desoxirribonucleases/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Plasmídeos/genética
5.
Nucleic Acids Res ; 40(8): 3443-55, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22189101

RESUMO

The exchange of genetic information between donor and acceptor DNA molecules by homologous recombination (HR) depends on the cleavage of phosphodiester bonds. Although double-stranded and single-stranded DNA breaks (SSBs) have both been invoked as triggers of HR, until very recently the focus has been primarily on the former type of DNA lesions mainly due to the paucity of SSB-based recombination models. Here, to investigate the role of nicked DNA molecules as HR-initiating substrates in human somatic cells, we devised a homology-directed gene targeting system based on exogenous donor and chromosomal target DNA containing recognition sequences for the adeno-associated virus sequence- and strand-specific endonucleases Rep78 and Rep68. We found that HR is greatly fostered if a SSB is not only introduced in the chromosomal acceptor but also in the donor DNA template. Our data are consistent with HR models postulating the occurrence of SSBs or single-stranded gaps in both donor and acceptor molecules during the genetic exchange process. These findings can guide the development of improved HR-based genome editing strategies in which sequence- and strand-specific endonucleolytic cleavage of the chromosomal target site is combined with that of the targeting vector.


Assuntos
Quebras de DNA de Cadeia Simples , Marcação de Genes , Recombinação Homóloga , Linhagem Celular , DNA/metabolismo , Endodesoxirribonucleases/metabolismo , Loci Gênicos , Humanos
6.
Nucleic Acids Res ; 37(17): 5725-36, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19651880

RESUMO

Homologous recombination (HR) is a highly accurate mechanism of DNA repair that can be exploited for homology-directed gene targeting. Since in most cell types HR occurs very infrequently (approximately 10(-6) to 10(-8)), its practical application has been largely restricted to specific experimental systems that allow selection of the few cells that become genetically modified. HR-mediated gene targeting has nonetheless revolutionized genetics by greatly facilitating the analysis of mammalian gene function. Recent studies showed that generation of double-strand DNA breaks at specific loci by designed endonucleases greatly increases the rate of homology-directed gene repair. These findings opened new perspectives for HR-based genome editing in higher eukaryotes. Here, we demonstrate by using donor DNA templates together with the adeno-associated virus (AAV) Rep78 and Rep68 proteins that sequence- and strand-specific cleavage at a native, predefined, human locus can also greatly enhance homology-directed gene targeting. Our findings argue for the development of other strategies besides direct induction of double-strand chromosomal breaks to achieve efficient and heritable targeted genetic modification of cells and organisms. Finally, harnessing the cellular HR pathway through Rep-mediated nicking expands the range of strategies that make use of AAV elements to bring about stable genetic modification of human cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases/metabolismo , Marcação de Genes/métodos , Recombinação Genética , Proteínas Virais/metabolismo , Cromossomos Humanos Par 19 , Células HeLa , Humanos , Transcrição Gênica
7.
J Control Release ; 317: 154-165, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31765703

RESUMO

Small, synthetic oligonucleotides (ON) are of great interest as potential disease modifying drugs, mainly because of their ability to modulate previously undruggable target mutations. To date, therapeutic applications of ON are, however, limited by their physicochemical properties, including poor stability, rapid excretion and low intracellular access. In order to overcome some of these shortcomings, ON are generally formulated using nanoparticle (NP) delivery systems. Alternatively, the poor stability can be circumvented by including chemical modifications to the backbone or sugars of the ON. Some of these modifications also result in better intracellular target access of these otherwise membrane-impermeable macromolecules. Therefore, complex formulation of ON into NP in order to overcome the hurdle of intracellular access might not always be needed, especially in case of local delivery. In this study, the delivery and functionality of chemically modified ON in free form was compared to polymeric NP assisted delivery, measuring their effectivity and efficiency. For this reason, phosphorothioate (PS) backbone-modified 18-mer ON with either 2'OMe or 2'MOE-modifications were selected, capable of eliciting exon-skipping of an aberrant exon in fluorescence based in vitro and in vivo model systems. The NP consisted of poly(D,L-lactic,co-glycolic acid) and poly-ß-amino-ester, previously demonstrated to successfully deliver nucleic acids via the pulmonary route. Several NP formulation parameters were tested in order to optimize the delivery of the ON, including ratio polymer:ON, NP size and concentration. The results reported here show clear differences between gymnotic and nanoparticle mediated ON delivery in terms of cellular uptake and local tissue distribution. In vitro, differences in exon-skipping efficiencies were observed with 2'OMe and 2'MOE ON either in free form or formulated in NP, with the striking observation that 2'OMe ON formulated in polymeric NP did not result in exon skipping. Gymnotic delivery of 2'MOE ON into the respiratory tract of mice resulted in functional delivery of exon-skipping ON into nasal epithelia and lungs as well as other downstream tissues and organs, pointing towards a gradual redistribution of locally delivered ONs, with limited but measurable systemic exposure. Conversely, NP-mediated delivery into the respiratory tract resulted in a more contained functional delivery at 10× lower ON doses compared to gymnotic delivery. Based on these findings we conclude that gymnotic delivery of 2'OMe or 2'MOE exon-skipping ON to the respiratory tract is effective, but that NP formulation might be advantageous in case spread of ON to non-target tissue can lead to undesired effects.


Assuntos
Nanopartículas , Ácidos Nucleicos , Animais , Camundongos , Oligonucleotídeos , RNA , Sistema Respiratório
8.
Mol Ther ; 16(4): 741-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18334989

RESUMO

Duchenne muscular dystrophy (DMD) is caused by mutations in the X chromosome-linked DMD gene, which encodes the sarcolemma-stabilizing protein-dystrophin. Initial attempts at DMD therapy deployed muscle progenitor cells from healthy donors. The utilization of these cells is, however, hampered by their immunogenicity, while those from DMD patients are scarce and display limited ex vivo replication. Nonmuscle cells with myogenic capacity may offer valuable alternatives especially if, to allow autologous transplantation, they are amenable to genetic intervention. As a paradigm for therapeutic gene transfer by heterotypic cell fusion we are investigating whether human mesenchymal stem cells (hMSCs) can serve as donors of recombinant DMD genes for recipient human muscle cells. Here, we show that forced MyoD expression in hMSCs greatly increases their tendency to participate in human myotube formation turning them into improved DNA delivery vehicles. Efficient loading of hMSCs with recombinant DMD was achieved through a new tropism-modified high-capacity adenoviral (hcAd) vector directing striated muscle-specific synthesis of full-length dystrophin. This study introduces the principle of genetic complementation of gene-defective cells via directed cell fusion and provides an initial framework to test whether transient MyoD synthesis in autologous, gene-corrected hMSCs increases their potential for treating DMD and, possibly, other muscular dystrophies.


Assuntos
Células-Tronco Mesenquimais/citologia , Células Musculares/metabolismo , Proteínas Musculares/biossíntese , Músculo Esquelético/metabolismo , Adenoviridae/genética , Fusão Celular/métodos , Células Cultivadas , Distrofina/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Células Musculares/citologia , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Proteína MyoD/biossíntese , Proteína MyoD/genética , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo
9.
Sci Rep ; 4: 5105, 2014 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-24870050

RESUMO

CRISPR/Cas9-derived RNA-guided nucleases (RGNs) are DNA targeting systems, which are rapidly being harnessed for gene regulation and gene editing purposes in model organisms and cell lines. As bona fide gene delivery vehicles, viral vectors may be particularly fit to broaden the applicability of RGNs to other cell types including dividing and quiescent primary cells. Here, the suitability of adenoviral vectors (AdVs) for delivering RGN components into various cell types is investigated. We demonstrate that AdVs, namely second-generation fiber-modified AdVs encoding Cas9 or single guide RNA (gRNA) molecules addressing the Cas9 nuclease to the AAVS1 "safe harbor" locus or to a recombinant model allele can be produced to high-titers (up to 20 × 10(10) transducing units/ml). Importantly, AdV-mediated transduction of gRNA:Cas9 ribonucleoprotein complexes into transformed and non-transformed cells yields rates of targeted mutagenesis similar to or approaching those achieved by isogenic AdVs encoding TALENs targeting the same AAVS1 chromosomal region. RGN-induced gene disruption frequencies in the various cell types ranged from 18% to 65%. We conclude that AdVs constitute a valuable platform for introducing RGNs into human somatic cells regardless of their transformation status. This approach should aid investigating the potential and limitations of RGNs in numerous experimental settings.


Assuntos
Adenoviridae/genética , Sistemas CRISPR-Cas/genética , Vetores Genéticos , RNA Guia de Cinetoplastídeos/genética , Terapia Genética , Humanos , Mutagênese
10.
Hum Gene Ther Methods ; 24(6): 399-411, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24059449

RESUMO

Zinc-finger nucleases (ZFNs) work as dimers to induce double-stranded DNA breaks (DSBs) at predefined chromosomal positions. In doing so, they constitute powerful triggers to edit and to interrogate the function of genomic sequences in higher eukaryotes. A preferred route to introduce ZFNs into somatic cells relies on their cotransduction with two integrase-defective lentiviral vectors (IDLVs) each encoding a monomer of a functional heterodimeric pair. The episomal nature of IDLVs diminishes the risk of genotoxicity and ensures the strict transient expression profile necessary to minimize deleterious effects associated with long-term ZFN activity. However, by deploying IDLVs and conventional lentiviral vectors encoding HPRT1- or eGFP-specific ZFNs, we report that DSB formation at target alleles is limited after IDLV-mediated ZFN transfer. This IDLV-specific underperformance stems, to a great extent, from the activity of chromatin-remodeling histone deacetylases (HDACs). Importantly, the prototypic and U.S. Food and Drug Administration-approved inhibitors of metal-dependent HDACs, trichostatin A and vorinostat, respectively, did not hinder illegitimate recombination-mediated repair of targeted chromosomal DSBs. This allowed rescuing IDLV-mediated site-directed mutagenesis to levels approaching those achieved by using their isogenic chromosomally integrating counterparts. Hence, HDAC inhibition constitutes an efficacious expedient to incorporate in genome-editing strategies based on transient IDLV-mediated ZFN expression. Finally, we compared two of the most commonly used readout systems to measure targeted gene knockout activities based on restriction and mismatch-sensitive endonucleases. These experiments indicate that these enzymatic assays display a similar performance.


Assuntos
Endodesoxirribonucleases/genética , Técnicas de Inativação de Genes/métodos , Vetores Genéticos/genética , Histona Desacetilases/metabolismo , Integrases/genética , Lentivirus/genética , Dedos de Zinco , Quebras de DNA de Cadeia Dupla , Endodesoxirribonucleases/química , Endodesoxirribonucleases/metabolismo , Células HEK293 , Células HeLa , Inibidores de Histona Desacetilases/farmacologia , Humanos , Hipoxantina Fosforribosiltransferase/genética , Células-Tronco Mesenquimais/metabolismo , Mioblastos/metabolismo
12.
PLoS One ; 5(6): e10954, 2010 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-20532169

RESUMO

Cell-to-cell fusion is involved in multiple fundamental biological processes. Prominent examples include osteoclast and giant cell formation, fertilization and skeletal myogenesis which involve macrophage, sperm-egg and myoblast fusion, respectively. Indeed, the importance of cell fusion is underscored by the wide range of homeostatic as well as pathologic processes in which it plays a key role. Therefore, rapid and sensitive systems to trace and measure cell fusion events in various experimental systems are in demand. Here, we introduce a bipartite cell fusion monitoring system based on a genetic switch responsive to the site-specific recombinase FLP. To allow flexible deployment in both dividing as well as non-dividing cell populations, inducer and reporter modules were incorporated in lentivirus vector particles. Moreover, the recombinase-inducible transcription units were designed in such a way as to minimize basal activity and chromosomal position effects in the "off" and "on" states, respectively. The lentivirus vector-based conditional gene expression assay was validated in primary human mesenchymal stem cells and in a differentiation model based on muscle progenitor cells from a Duchenne muscular dystrophy patient using reporter genes compatible with live- and single-cell imaging and with whole population measurements. Using the skeletal muscle cell differentiation model, we showed that the new assay displays low background activity, a 2-log dynamic range, high sensitivity and is amenable to the investigation of cell fusion kinetics. The utility of the bipartite cell fusion monitoring system was underscored by a study on the impact of drug- and RNAi-mediated p38 MAPK inhibition on human myocyte differentiation. Finally, building on the capacity of lentivirus vectors to readily generate transgenic animals the present FLP-inducible system should be adaptable, alone or together with Cre/loxP-based assays, to cell lineage tracing and conditional gene manipulation studies in vivo.


Assuntos
Fusão Celular , Expressão Gênica , Vetores Genéticos , Lentivirus/genética , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/metabolismo
13.
PLoS One ; 3(8): e3084, 2008 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-18769728

RESUMO

A prominent goal in gene therapy research concerns the development of gene transfer vehicles that can integrate exogenous DNA at specific chromosomal loci to prevent insertional oncogenesis and provide for long-term transgene expression. Adenovirus (Ad) vectors arguably represent the most efficient delivery systems of episomal DNA into eukaryotic cell nuclei. The most advanced recombinant Ads lack all adenoviral genes. This renders these so-called high-capacity (hc) Ad vectors less cytotoxic/immunogenic than those only deleted in early regions and creates space for the insertion of large/multiple transgenes. The versatility of hcAd vectors is been increased by capsid modifications to alter their tropism and by the incorporation into their genomes of sequences promoting chromosomal insertion of exogenous DNA. Adeno-associated virus (AAV) can insert its genome into a specific human locus designated AAVS1. Trans- and cis-acting elements needed for this reaction are the AAV Rep78/68 proteins and Rep78/68-binding sequences, respectively. Here, we describe the generation, characterization and testing of fiber-modified dual hcAd/AAV hybrid vectors (dHVs) containing both these elements. Due to the inhibitory effects of Rep78/68 on Ad-dependent DNA replication, we deployed a recombinase-inducible gene switch to repress Rep68 synthesis during vector rescue and propagation. Flow cytometric analyses revealed that rep68-positive dHVs can be produced similarly well as rep68-negative control vectors. Western blot experiments and immunofluorescence microscopy analyses demonstrated transfer of recombinase-dependent rep68 genes into target cells. Studies in HeLa cells and in the dystrophin-deficient myoblasts from a Duchenne muscular dystrophy (DMD) patient showed that induction of Rep68 synthesis in cells transduced with fiber-modified and rep68-positive dHVs leads to increased stable transduction levels and AAVS1-targeted integration of vector DNA. These results warrant further investigation especially considering the paucity of vector systems allowing permanent phenotypic correction of patient-own cell types with large DNA (e.g. recombinant full-length DMD genes).


Assuntos
Cromossomos Humanos/genética , Elementos de DNA Transponíveis/genética , Genoma Humano , Adenoviridae/genética , Animais , Cromossomos/genética , Replicação do DNA , Terapia Genética/métodos , Vetores Genéticos , Humanos , Modelos Animais
14.
Mol Ther ; 13(5): 976-86, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16443396

RESUMO

Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene (DMD), making it amenable to gene- or cell-based therapies. Another possible treatment entails the combination of both principles by transplantation of autologous myogenic cells after their genetic complementation. This approach requires efficient and stable transduction of these cells with recombinant DMD. Recently, we generated a dual high-capacity (hc) adenovirus (Ad)-adeno-associated virus (AAV) hybrid vector (HV) that can deliver two full-length dystrophin-encoding modules into target cells. We showed that HV transduction of human cells containing AAV Rep proteins leads to the insertion of foreign DNA into the AAVS1 locus. Here, we improved HV entry into muscle cells from DMD patients. After having verified that these cells barely express the coxsackie B virus and Ad receptor (CAR), which constitutes the attachment molecule for Ad serotype 5 (Ad5) fibers, we equipped dual hcAd/AAV HV particles with Ad serotype 50 fiber domains to achieve CAR-independent uptake. These retargeted vectors complemented much more efficiently the genetic defect of dystrophin-defective myoblasts and myotubes than their isogenic counterparts with conventional Ad5 fibers. Importantly, the accumulation of beta-dystroglycan along the membranes of vector-treated DMD myotubes indicated proper assembly of dystrophin-associated glycoprotein complexes.


Assuntos
Distrofina/biossíntese , Distrofina/genética , Vetores Genéticos , Células Musculares/metabolismo , Transdução Genética , Adenoviridae/classificação , Adenoviridae/genética , DNA Recombinante/genética , Dependovirus/genética , Técnicas de Transferência de Genes , Terapia Genética , Células HeLa , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Sorotipagem , Integração Viral
15.
Hum Mol Genet ; 15(2): 213-21, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16321987

RESUMO

Duchenne muscular dystrophy (DMD) is the most prevalent inheritable muscle disease. It is caused by mutations in the approximately 2.5-megabase dystrophin (Dys) encoding gene. Therapeutic attempts at DMD have relied on injection of allogeneic Dys-positive myoblasts. The immune rejection of these cells and their limited availability have prompted the search for alternative therapies and sources of myogenic cells. Stem cell-based gene therapy aims to restore tissue function by the transplantation of gene-corrected autologous cells. It depends on (i) the capacity of stem cells to participate in tissue regeneration and (ii) the efficient genetic correction of defective autologous stem cells. We explored the potential of bone marrow-derived human mesenchymal stem cells (hMSCs) genetically modified with the full-length Dys-coding sequence to engage in myogenesis. By tagging hMSCs with enhanced green fluorescent protein (EGFP) or the membrane dye PKH26, we demonstrated that they could participate in myotube formation when cultured together with differentiating human myoblasts. Experiments performed with EGFP-marked hMSCs and DsRed-labeled DMD myoblasts revealed that the EGFP-positive DMD myotubes were also DsRed-positive indicating that hMSCs participate in human myogenesis through cellular fusion. Finally, we showed that hMSCs transduced with a tropism-modified high-capacity hybrid viral vector encoding full-length Dys could complement the genetic defect of DMD myotubes.


Assuntos
Distrofina/metabolismo , Terapia Genética/métodos , Células-Tronco Mesenquimais/metabolismo , Desenvolvimento Muscular/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Distrofia Muscular de Duchenne/terapia , Transplante de Células-Tronco , Fusão Celular , Vetores Genéticos/uso terapêutico , Proteínas de Fluorescência Verde , Humanos , Imunofenotipagem , Lentivirus , Proteínas Luminescentes , Desenvolvimento Muscular/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa