Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nature ; 511(7507): 86-9, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24870238

RESUMEN

In female mice, two forms of X-chromosome inactivation (XCI) ensure the selective silencing of female sex chromosomes during mouse embryogenesis. Beginning at the four-cell stage, imprinted XCI (iXCI) exclusively silences the paternal X chromosome. Later, around implantation, epiblast cells of the inner cell mass that give rise to the embryo reactivate the paternal X chromosome and undergo a random form of XCI (rXCI). Xist, a long non-coding RNA crucial for both forms of XCI, is activated by the ubiquitin ligase RLIM (also known as Rnf12). Although RLIM is required for triggering iXCI in mice, its importance for rXCI has been controversial. Here we show that RLIM levels are downregulated in embryonic cells undergoing rXCI. Using mouse genetics we demonstrate that female cells lacking RLIM from pre-implantation stages onwards show hallmarks of XCI, including Xist clouds and H3K27me3 foci, and have full embryogenic potential. These results provide evidence that RLIM is dispensable for rXCI, indicating that in mice an RLIM-independent mechanism activates Xist in the embryo proper.


Asunto(s)
Estratos Germinativos/embriología , Estratos Germinativos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Inactivación del Cromosoma X/genética , Animales , Regulación hacia Abajo , Implantación del Embrión , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Histonas/química , Histonas/metabolismo , Hibridación Fluorescente in Situ , Lisina/metabolismo , Metilación , Ratones , Ratones Noqueados , ARN Largo no Codificante/genética , Ubiquitina-Proteína Ligasas/genética
2.
Nature ; 467(7318): 977-81, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20962847

RESUMEN

Two forms of X-chromosome inactivation (XCI) ensure the selective silencing of female sex chromosomes during mouse embryogenesis. Imprinted XCI begins with the detection of Xist RNA expression on the paternal X chromosome (Xp) at about the four-cell stage of embryonic development. In the embryonic tissues of the inner cell mass, a random form of XCI occurs in blastocysts that inactivates either Xp or the maternal X chromosome (Xm). Both forms of XCI require the non-coding Xist RNA that coats the inactive X chromosome from which it is expressed. Xist has crucial functions in the silencing of X-linked genes, including Rnf12 (refs 3, 4) encoding the ubiquitin ligase RLIM (RING finger LIM-domain-interacting protein). Here we show, by targeting a conditional knockout of Rnf12 to oocytes where RLIM accumulates to high levels, that the maternal transmission of the mutant X chromosome (Δm) leads to lethality in female embryos as a result of defective imprinted XCI. We provide evidence that in Δm female embryos the initial formation of Xist clouds and Xp silencing are inhibited. In contrast, embryonic stem cells lacking RLIM are able to form Xist clouds and silence at least some X-linked genes during random XCI. These results assign crucial functions to the maternal deposit of Rnf12/RLIM for the initiation of imprinted XCI.


Asunto(s)
Cromosomas de los Mamíferos/genética , Impresión Genómica , Madres , Proteínas Represoras/metabolismo , Inactivación del Cromosoma X/genética , Cromosoma X/genética , Animales , Animales Congénicos , Blastocisto/metabolismo , Línea Celular , Pérdida del Embrión/genética , Padre , Femenino , Silenciador del Gen , Masculino , Ratones , Ratones Transgénicos , ARN Largo no Codificante , ARN no Traducido/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Ubiquitina-Proteína Ligasas
3.
Sci Rep ; 8(1): 8811, 2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29891904

RESUMEN

The characteristic six layers of the mammalian neocortex develop sequentially as neurons are generated by neural progenitors and subsequently migrate past older neurons to their final position in the cortical plate. One of the earliest steps of neuronal differentiation is the formation of an axon. Small GTPases play essential roles during this process by regulating cytoskeletal dynamics and intracellular trafficking. While the function of GTPases has been studied extensively in cultured neurons and in vivo much less is known about their upstream regulators. Here we show that Arhgef7 (also called ßPix or Cool1) is essential for axon formation during cortical development. The loss of Arhgef7 results in an extensive loss of axons in cultured neurons and in the developing cortex. Arhgef7 is a guanine-nucleotide exchange factor (GEF) for Cdc42, a GTPase that has a central role in directing the formation of axons during brain development. However, active Cdc42 was not able to rescue the knockdown of Arhgef7. We show that Arhgef7 interacts with the GTPase TC10 that is closely related to Cdc42. Expression of active TC10 can restore the ability to extend axons in Arhgef7-deficient neurons. Our results identify an essential role of Arhgef7 during neuronal development that promotes axon formation upstream of TC10.


Asunto(s)
Axones/fisiología , Diferenciación Celular , Corteza Cerebral/embriología , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Factores de Intercambio de Guanina Nucleótido Rho/deficiencia , Proteína de Unión al GTP cdc42/metabolismo
4.
Mech Dev ; 117(1-2): 75-85, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12204249

RESUMEN

The crucial involvement of CLIM/NLI/Ldb cofactors for the exertion of the biological activity of LIM homeodomain transcription factors (LIM-HD) has been demonstrated. In this paper we show that CLIM cofactors are widely expressed during zebrafish development with high protein levels in specific neuronal cell types where LIM-HD proteins of the Isl class are synthesized. The overexpression of a dominant-negative CLIM molecule (DN-CLIM) that contains the LIM interaction domain (LID) during early developmental stages of zebrafish embryos results in an impairment of eye and midbrain-hindbrain boundary (MHB) development and disturbances in the formation of the anterior midline. On a cellular level we show that the outgrowth of peripheral but not central axons from Rohon Beard (RB) and trigeminal sensory neurons is inhibited by DN-CLIM overexpression. We demonstrate a further critical role of CLIM cofactors for axonal outgrowth of motor neurons. Additionally, DN-CLIM overexpression causes an increase of Isl-protein expression levels in specific neuronal cell types, likely due to a protection of the DN-CLIM/LIM-HD complex from proteasomal degradation. Our results demonstrate multiple roles of the CLIM cofactor family for the development of entire organs, axonal outgrowth of specific neurons and protein expression levels.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Axones/metabolismo , Axones/ultraestructura , Encéfalo/embriología , Encéfalo/metabolismo , Ojo/embriología , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/genética , Inmunohistoquímica , Hibridación in Situ , Neuronas/metabolismo , Factores de Transcripción/genética , Pez Cebra/genética
5.
Proc Natl Acad Sci U S A ; 104(38): 15000-5, 2007 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-17848518

RESUMEN

Complexes composed of multiple proteins regulate most cellular functions. However, our knowledge about the molecular mechanisms governing the assembly and dynamics of these complexes in cells remains limited. The in vivo activity of LIM homeodomain (LIM-HD) proteins, a class of transcription factors that regulates neuronal development, depends on the high-affinity association of their LIM domains with cofactor of LIM homeodomain proteins (LIM-HDs) (CLIM, also known as Ldb or NLI). CLIM cofactors recruit single-stranded DNA-binding protein 1 (SSDP1, also known as SSBP3), and this interaction is important for the activation of the LIM-HD/CLIM protein complex in vivo. Here, we identify a cascade of specific protein interactions that protect LIM-HD multiprotein complexes from proteasomal degradation. In this cascade, CLIM stabilizes LIM-HDs, and SSDP1 stabilizes CLIM. Furthermore, we show that stabilizing cofactors prevent binding of ubiquitin ligases to multiple protein interaction domains in LIM-HD recruited protein complexes. Together, our results indicate a combinatorial code that selects specific multiprotein complexes via proteasomal degradation in cells with broad implications for the assembly and specificity of multiprotein complexes.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Complejos Multiproteicos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción/metabolismo , Células Cultivadas , Modelos Biológicos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transfección
6.
Genes Dev ; 19(19): 2307-19, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16204183

RESUMEN

LIM kinase 1 (LIMK1) controls important cellular functions such as morphogenesis, cell motility, tumor cell metastasis, development of neuronal projections, and growth cone actin dynamics. We have investigated the role of the RING finger protein Rnf6 during neuronal development and detected high Rnf6 protein levels in developing axonal projections of motor and DRG neurons during mouse embryogenesis as well as cultured hippocampal neurons. RNAi-mediated knock-down experiments in primary hippocampal neurons identified Rnf6 as a regulator of axon outgrowth. Consistent with a role in axonal growth, we found that Rnf6 binds to, polyubiquitinates, and targets LIMK1 for proteasomal degradation in growth cones of primary hippocampal neurons. Rnf6 is functionally linked to LIMK1 during the development of axons, as the changes in axon outgrowth induced by up- or down-regulation of Rnf6 levels can be restored by modulation of LIMK1 expression. Thus, these results assign a specific role for Rnf6 in the control of cellular LIMK1 concentrations and indicate a new function for the ubiquitin/proteasome system in regulating local growth cone actin dynamics.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Conos de Crecimiento/enzimología , Hipocampo/embriología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Quinasas/metabolismo , Actinas/metabolismo , Animales , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Cricetulus , Proteínas de Unión al ADN/genética , Hipocampo/citología , Humanos , Quinasas Lim , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética , Ubiquitina/metabolismo
7.
Nature ; 416(6876): 99-103, 2002 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-11882901

RESUMEN

The interactions of distinct cofactor complexes with transcription factors are decisive determinants for the regulation of gene expression. Depending on the bound cofactor, transcription factors can have either repressing or transactivating activities. To allow a switch between these different states, regulated cofactor exchange has been proposed; however, little is known about the molecular mechanisms that are involved in this process. LIM homeodomain (LIM-HD) transcription factors associate with RLIM (RING finger LIM domain-binding protein) and with CLIM (cofactor of LIM-HD proteins; also known as NLI, Ldb and Chip) cofactors. The co-repressor RLIM inhibits the function of LIM-HD transcription factors, whereas interaction with CLIM proteins is important for the exertion of the biological activity conferred by LIM-HD transcription-factors. Here we identify RLIM as a ubiquitin protein ligase that is able to target CLIM cofactors for degradation through the 26S proteasome pathway. Furthermore, we demonstrate a ubiquitination-dependent association of RLIM with LIM-HD proteins in the presence of CLIM cofactors. Our data provide a mechanistic basis for cofactor exchange on DNA-bound transcription factors, and probably represent a general mechanism of transcriptional regulation.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Péptido Hidrolasas/metabolismo , Complejo de la Endopetidasa Proteasomal , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células CHO , Línea Celular , Cricetinae , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Proteínas con Dominio LIM , Ligasas/metabolismo , Metaloproteínas/metabolismo , Ratones , Unión Proteica , Proteínas Proto-Oncogénicas , Transfección , Ubiquitina-Proteína Ligasas
8.
Methods Cell Sci ; 25(1-2): 85-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14739592

RESUMEN

The stabilities of many key proteins are regulated, e.g. via ubiquitination and proteasomal degradation, with important biological consequences. We present a convenient method that allows the analysis and comparison of protein stabilities during embryogenesis using early zebrafish development as a model system. Basically, this method involves ectopic overexpression of epitope-tagged proteins via mRNA injections in one-to-four-cell stage embryos and subsequent protein detection after various time points. Indeed, the protein stability of the ubiquitin ligase RLIM, which is able to autoubiquitinate and target itself for proteasomal degradation, was much shorter when compared to a protein consisting of a Myc epitope-tag and a nuclear localization domain. Thus, this method may be used more widely for the study of developmental protein stability.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Estabilidad del ARN/fisiología , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Pez Cebra/metabolismo , Animales , Clonación Molecular , Embrión no Mamífero/metabolismo , Microinyecciones , Desnaturalización Proteica , Pez Cebra/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA