Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 8(1): 17348, 2018 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-30478432

RESUMEN

The inner ear is a complex structure responsible for hearing and balance, and organ pathology is associated with deafness and balance disorders. To evaluate the role of epigenomic dynamics, we performed whole genome bisulfite sequencing at key time points during the development and maturation of the mouse inner ear sensory epithelium (SE). Our single-nucleotide resolution maps revealed variations in both general characteristics and dynamics of DNA methylation over time. This allowed us to predict the location of non-coding regulatory regions and to identify several novel candidate regulatory factors, such as Bach2, that connect stage-specific regulatory elements to molecular features that drive the development and maturation of the SE. Constructing in silico regulatory networks around sites of differential methylation enabled us to link key inner ear regulators, such as Atoh1 and Stat3, to pathways responsible for cell lineage determination and maturation, such as the Notch pathway. We also discovered that a putative enhancer, defined as a low methylated region (LMR), can upregulate the GJB6 gene and a neighboring non-coding RNA. The study of inner ear SE methylomes revealed novel regulatory regions in the hearing organ, which may improve diagnostic capabilities, and has the potential to guide the development of therapeutics for hearing loss by providing multiple intervention points for manipulation of the auditory system.


Asunto(s)
Conexina 30/genética , Metilación de ADN/fisiología , Oído Interno/embriología , Oído Interno/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Sordera/genética , Oído Interno/citología , Elementos de Facilitación Genéticos , Epitelio/embriología , Epitelio/crecimiento & desarrollo , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Factores del Dominio POU/genética , Embarazo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
2.
Cell Rep ; 20(6): 1448-1462, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28793267

RESUMEN

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.


Asunto(s)
Células Madre Embrionarias/citología , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Defectos del Tubo Neural/genética , Tubo Neural/embriología , Línea Celular , Metilación de ADN , Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis
3.
Stem Cell Reports ; 9(3): 999-1015, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28844656

RESUMEN

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.


Asunto(s)
Células de la Médula Ósea/metabolismo , Metilación de ADN/genética , Células Madre Mesenquimatosas/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Edad , Anciano , Anciano de 80 o más Años , Sitios de Unión , Células Cultivadas , Islas de CpG/genética , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Genoma Humano , Humanos , Persona de Mediana Edad , Análisis de Secuencia de ADN , Factores de Transcripción/metabolismo , Adulto Joven
5.
Cell Rep ; 19(2): 281-294, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28402852

RESUMEN

Activating germline mutations in STAT3 were recently identified as a cause of neonatal diabetes mellitus associated with beta-cell autoimmunity. We have investigated the effect of an activating mutation, STAT3K392R, on pancreatic development using induced pluripotent stem cells (iPSCs) derived from a patient with neonatal diabetes and pancreatic hypoplasia. Early pancreatic endoderm differentiated similarly from STAT3K392R and healthy-control cells, but in later stages, NEUROG3 expression was upregulated prematurely in STAT3K392R cells together with insulin (INS) and glucagon (GCG). RNA sequencing (RNA-seq) showed robust NEUROG3 downstream targets upregulation. STAT3 mutation correction with CRISPR/Cas9 reversed completely the disease phenotype. STAT3K392R-activating properties were not explained fully by altered DNA-binding affinity or increased phosphorylation. Instead, reporter assays demonstrated NEUROG3 promoter activation by STAT3 in pancreatic cells. Furthermore, proteomic and immunocytochemical analyses revealed increased nuclear translocation of STAT3K392R. Collectively, our results demonstrate that the STAT3K392R mutation causes premature endocrine differentiation through direct induction of NEUROG3 expression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Diferenciación Celular/genética , Diabetes Mellitus/genética , Proteínas del Tejido Nervioso/biosíntesis , Factor de Transcripción STAT3/genética , Autoinmunidad/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sistemas CRISPR-Cas , Línea Celular , Diabetes Mellitus/etiología , Diabetes Mellitus/patología , Regulación del Desarrollo de la Expresión Génica , Glucagón/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Mutación , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas , Factor de Transcripción STAT3/biosíntesis
6.
Nat Commun ; 8: 14454, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28195574

RESUMEN

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.


Asunto(s)
Sistemas CRISPR-Cas/genética , Distrofina/genética , Edición Génica/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatología , Animales , Proteínas Bacterianas/genética , Proteína 9 Asociada a CRISPR , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Endonucleasas/genética , Terapia Genética/métodos , Vectores Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/terapia , Mutación , Miocardio , Enfermedades Neuromusculares/terapia , ARN Guía de Kinetoplastida , Eliminación de Secuencia
7.
Stem Cell Reports ; 6(2): 200-12, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26777058

RESUMEN

Reports on the retention of somatic cell memory in induced pluripotent stem cells (iPSCs) have complicated the selection of the optimal cell type for the generation of iPSC biobanks. To address this issue we compared transcriptomic, epigenetic, and differentiation propensities of genetically matched human iPSCs derived from fibroblasts and blood, two tissues of the most practical relevance for biobanking. Our results show that iPSC lines derived from the same donor are highly similar to each other. However, genetic variation imparts a donor-specific expression and methylation profile in reprogrammed cells that leads to variable functional capacities of iPSC lines. Our results suggest that integration-free, bona fide iPSC lines from fibroblasts and blood can be combined in repositories to form biobanks. Due to the impact of genetic variation on iPSC differentiation, biobanks should contain cells from large numbers of donors.


Asunto(s)
Diferenciación Celular/genética , Variación Genética , Células Madre Pluripotentes Inducidas/citología , Bancos de Muestras Biológicas , Metilación de ADN/genética , Epigénesis Genética , Células Eritroides/citología , Femenino , Fibroblastos/metabolismo , Hematopoyesis/genética , Humanos , Masculino , Donantes de Tejidos , Transcripción Genética
8.
BMC Genomics ; 16: 1083, 2015 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-26692029

RESUMEN

BACKGROUND: ChIP-seq is highly utilized for mapping histone modifications that are informative about gene regulation and genome annotations. For example, applying ChIP-seq to histone modifications such as H3K4me1 has facilitated generating epigenomic maps of putative enhancers. This powerful technology, however, is limited in its application by the large number of cells required. ChIP-seq involves extensive manipulation of sample material and multiple reactions with limited quality control at each step, therefore, scaling down the number of cells required has proven challenging. Recently, several methods have been proposed to overcome this limit but most of these methods require extensive optimization to tailor the protocol to the specific antibody used or number of cells being profiled. RESULTS: Here we describe a robust, yet facile method, which we named carrier ChIP-seq (cChIP-seq), for use on limited cell amounts. cChIP-seq employs a DNA-free histone carrier in order to maintain the working ChIP reaction scale, removing the need to tailor reactions to specific amounts of cells or histone modifications to be assayed. We have applied our method to three different histone modifications, H3K4me3, H3K4me1 and H3K27me3 in the K562 cell line, and H3K4me1 in H1 hESCs. We successfully obtained epigenomic maps for these histone modifications starting with as few as 10,000 cells. We compared cChIP-seq data to data generated as part of the ENCODE project. ENCODE data are the reference standard in the field and have been generated starting from tens of million of cells. Our results show that cChIP-seq successfully recapitulates bulk data. Furthermore, we showed that the differences observed between small-scale ChIP-seq data and ENCODE data are largely to be due to lab-to-lab variability rather than operating on a reduced scale. CONCLUSIONS: Data generated using cChIP-seq are equivalent to reference epigenomic maps from three orders of magnitude more cells. Our method offers a robust and straightforward approach to scale down ChIP-seq to as low as 10,000 cells. The underlying principle of our strategy makes it suitable for being applied to a vast range of chromatin modifications without requiring expensive optimization. Furthermore, our strategy of a DNA-free carrier can be adapted to most ChIP-seq protocols.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Código de Histonas , Análisis de Secuencia de ADN/métodos , Línea Celular , Epigenómica/métodos , Humanos , Células K562
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...