Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 25(9): 104954, 2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-36093058

RESUMEN

Regulation of chromatin accessibility is critical for cell fate decisions. Chromatin structure responds to extrinsic environments rapidly. The traditional adult stem cell isolation approach requires tissue dissociation, which triggers stem cell activation and leads to alterations in chromatin structure. To preserve the in vivo chromatin states, we utilized the PFA-perfusion-based isolation approach and characterized the DNA regulatory landscapes during muscle stem cell quiescence exit and aging. We showed that aged SCs display a chronically activated chromatin signature. Detailed analysis of the chromatin accessibility profiles identified key enhancer elements for SC quiescence. Constant activation of the enhancer elements promotes stemness and prevents SCs from differentiation, whereas genetic deletion causes cell-cycle arrest and leads to defects in activation. Our comprehensive characterization of the chromatin accessibility and transcriptomic landscapes in SC quiescence and aging broadens our understanding of these processes and identifies key distal regulatory elements for SC function.

2.
Proc Natl Acad Sci U S A ; 117(51): 32464-32475, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33293420

RESUMEN

Epigenetics regulation plays a critical role in determining cell identity by controlling the accessibility of lineage-specific regulatory regions. In muscle stem cells, epigenetic mechanisms of how chromatin accessibility is modulated during cell fate determination are not fully understood. Here, we identified a long noncoding RNA, LncMyoD, that functions as a chromatin modulator for myogenic lineage determination and progression. The depletion of LncMyoD in muscle stem cells led to the down-regulation of myogenic genes and defects in myogenic differentiation. LncMyoD exclusively binds with MyoD and not with other myogenic regulatory factors and promotes transactivation of target genes. The mechanistic study revealed that loss of LncMyoD prevents the establishment of a permissive chromatin environment at myogenic E-box-containing regions, therefore restricting the binding of MyoD. Furthermore, the depletion of LncMyoD strongly impairs the reprogramming of fibroblasts into the myogenic lineage. Taken together, our study shows that LncMyoD associates with MyoD and promotes myogenic gene expression through modulating MyoD accessibility to chromatin, thereby regulating myogenic lineage determination and progression.


Asunto(s)
Cromatina/genética , ARN Largo no Codificante/genética , Células Satélite del Músculo Esquelético/fisiología , Animales , Diferenciación Celular/genética , Linaje de la Célula , Transdiferenciación Celular , Cromatina/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Desarrollo de Músculos/fisiología , Proteína MioD/genética , Mioblastos/citología , Mioblastos/fisiología , Células Satélite del Músculo Esquelético/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA