Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 20(6): 1448-1462, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28793267

RESUMO

We currently lack a comprehensive understanding of the mechanisms underlying neural tube formation and their contributions to neural tube defects (NTDs). Developing a model to study such a complex morphogenetic process, especially one that models human-specific aspects, is critical. Three-dimensional, human embryonic stem cell (hESC)-derived neural rosettes (NRs) provide a powerful resource for in vitro modeling of human neural tube formation. Epigenomic maps reveal enhancer elements unique to NRs relative to 2D systems. A master regulatory network illustrates that key NR properties are related to their epigenomic landscapes. We found that folate-associated DNA methylation changes were enriched within NR regulatory elements near genes involved in neural tube formation and metabolism. Our comprehensive regulatory maps offer insights into the mechanisms by which folate may prevent NTDs. Lastly, our distal regulatory maps provide a better understanding of the potential role of neurological-disorder-associated SNPs.


Assuntos
Células-Tronco Embrionárias/citologia , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Defeitos do Tubo Neural/genética , Tubo Neural/embriologia , Linhagem Celular , Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Elementos Facilitadores Genéticos , Humanos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese
2.
Stem Cell Reports ; 9(3): 999-1015, 2017 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-28844656

RESUMO

Human bone marrow stromal cells, or mesenchymal stem cells (BM-MSCs), need expansion prior to use as cell-based therapies in immunological and tissue repair applications. Aging and expansion of BM-MSCs induce epigenetic changes that can impact therapeutic outcomes. By applying sequencing-based methods, we reveal that the breadth of DNA methylation dynamics associated with aging and expansion is greater than previously reported. Methylation changes are enriched at known distal transcription factor binding sites such as enhancer elements, instead of CpG-rich regions, and are associated with changes in gene expression. From this, we constructed hypo- and hypermethylation-specific regulatory networks, including a sub-network of BM-MSC master regulators and their predicted target genes, and identified putatively disrupted signaling pathways. Our genome-wide analyses provide a broader overview of age- and expansion-induced DNA methylation changes and a better understanding of the extent to which these changes alter gene expression and functionality of human BM-MSCs.


Assuntos
Células da Medula Óssea/metabolismo , Metilação de DNA/genética , Células-Tronco Mesenquimais/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Sítios de Ligação , Células Cultivadas , Ilhas de CpG/genética , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Genoma Humano , Humanos , Pessoa de Meia-Idade , Análise de Sequência de DNA , Fatores de Transcrição/metabolismo , Adulto Jovem
3.
Nat Commun ; 8: 14454, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28195574

RESUMO

Gene replacement therapies utilizing adeno-associated viral (AAV) vectors hold great promise for treating Duchenne muscular dystrophy (DMD). A related approach uses AAV vectors to edit specific regions of the DMD gene using CRISPR/Cas9. Here we develop multiple approaches for editing the mutation in dystrophic mdx4cv mice using single and dual AAV vector delivery of a muscle-specific Cas9 cassette together with single-guide RNA cassettes and, in one approach, a dystrophin homology region to fully correct the mutation. Muscle-restricted Cas9 expression enables direct editing of the mutation, multi-exon deletion or complete gene correction via homologous recombination in myogenic cells. Treated muscles express dystrophin in up to 70% of the myogenic area and increased force generation following intramuscular delivery. Furthermore, systemic administration of the vectors results in widespread expression of dystrophin in both skeletal and cardiac muscles. Our results demonstrate that AAV-mediated muscle-specific gene editing has significant potential for therapy of neuromuscular disorders.


Assuntos
Sistemas CRISPR-Cas/genética , Distrofina/genética , Edição de Genes/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatologia , Animais , Proteínas de Bactérias/genética , Proteína 9 Associada à CRISPR , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Dependovirus/genética , Modelos Animais de Doenças , Endonucleases/genética , Terapia Genética/métodos , Vetores Genéticos , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/terapia , Mutação , Miocárdio , Doenças Neuromusculares/terapia , RNA Guia de Cinetoplastídeos , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA