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

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Cell ; 165(2): 382-95, 2016 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-27040500

RESUMEN

Gene duplication is a major evolutionary force driving adaptation and speciation, as it allows for the acquisition of new functions and can augment or diversify existing functions. Here, we report a gene duplication event that yielded another outcome--the generation of antagonistic functions. One product of this duplication event--UPF3B--is critical for the nonsense-mediated RNA decay (NMD) pathway, while its autosomal counterpart--UPF3A--encodes an enigmatic protein previously shown to have trace NMD activity. Using loss-of-function approaches in vitro and in vivo, we discovered that UPF3A acts primarily as a potent NMD inhibitor that stabilizes hundreds of transcripts. Evidence suggests that UPF3A acquired repressor activity through simple impairment of a critical domain, a rapid mechanism that may have been widely used in evolution. Mice conditionally lacking UPF3A exhibit "hyper" NMD and display defects in embryogenesis and gametogenesis. Our results support a model in which UPF3A serves as a molecular rheostat that directs developmental events.


Asunto(s)
Desarrollo Embrionario , Genes Duplicados , Degradación de ARNm Mediada por Codón sin Sentido , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular Tumoral , Evolución Molecular , Gametogénesis , Células HeLa , Humanos , Ratones
2.
Cell Rep ; 17(1): 149-164, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27681428

RESUMEN

The developmental origins of most adult stem cells are poorly understood. Here, we report the identification of a transcription factor-RHOX10-critical for the initial establishment of spermatogonial stem cells (SSCs). Conditional loss of the entire 33-gene X-linked homeobox gene cluster that includes Rhox10 causes progressive spermatogenic decline, a phenotype indistinguishable from that caused by loss of only Rhox10. We demonstrate that this phenotype results from dramatically reduced SSC generation. By using a battery of approaches, including single-cell-RNA sequencing (scRNA-seq) analysis, we show that Rhox10 drives SSC generation by promoting pro-spermatogonia differentiation. Rhox10 also regulates batteries of migration genes and promotes the migration of pro-spermatogonia into the SSC niche. The identification of an X-linked homeobox gene that drives the initial generation of SSCs has implications for the evolution of X-linked gene clusters and sheds light on regulatory mechanisms influencing adult stem cell generation in general.


Asunto(s)
Células Madre Germinales Adultas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Ligados a X , Proteínas de Homeodominio/genética , Espermatogénesis/genética , Espermatogonias/metabolismo , Células Madre Germinales Adultas/citología , Animales , Genes del Desarrollo , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Noqueados , Familia de Multigenes , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Espermatogonias/citología
3.
PLoS One ; 10(3): e0118549, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25790000

RESUMEN

RNA interference (RNAi) is widely used to determine the function of genes. We chose this approach to assess the collective function of the highly related reproductive homeobox 3 (Rhox3) gene paralogs. Using a Rhox3 short hairpin (sh) RNA with 100% complementarity to all 8 Rhox3 paralogs, expressed from a CRE-regulated transgene, we successfully knocked down Rhox3 expression in male germ cells in vivo. These Rhox3-shRNA transgenic mice had dramatic defects in spermatogenesis, primarily in spermatocytes and round spermatids. To determine whether this phenotype was caused by reduced Rhox3 expression, we generated mice expressing the Rhox3-shRNA but lacking the intended target of the shRNA-Rhox3. These double-mutant mice had a phenotype indistinguishable from Rhox3-shRNA-expressing mice that was different from mice lacking the Rhox3 paralogs, indicating that the Rhox3 shRNA disrupts spermatogenesis independently of Rhox3. Rhox3-shRNA transgenic mice displayed few alterations in the expression of protein-coding genes, but instead exhibited reduced levels of all endogenous siRNAs we tested. This supported a model in which the Rhox3 shRNA causes spermatogenic defects by sequestering one or more components of the endogenous small RNA biogenesis machinery. Our study serves as a warning for those using shRNA approaches to investigate gene functions in vivo.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Modelos Biológicos , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Espermatogénesis/genética , Animales , Cartilla de ADN/genética , Técnicas de Silenciamiento del Gen/métodos , Células HeLa , Humanos , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Transgénicos , Análisis por Micromatrices
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA