Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
Cell ; 141(6): 956-69, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20550932

RESUMEN

During X chromosome inactivation (XCI), Xist RNA coats and silences one of the two X chromosomes in female cells. Little is known about how XCI spreads across the chromosome, although LINE-1 elements have been proposed to play a role. Here we show that LINEs participate in creating a silent nuclear compartment into which genes become recruited. A subset of young LINE-1 elements, however, is expressed during XCI, rather than being silenced. We demonstrate that such LINE expression requires the specific heterochromatic state induced by Xist. These LINEs often lie within escape-prone regions of the X chromosome, but close to genes that are subject to XCI, and are associated with putative endo-siRNAs. LINEs may thus facilitate XCI at different levels, with silent LINEs participating in assembly of a heterochromatic nuclear compartment induced by Xist, and active LINEs participating in local propagation of XCI into regions that would otherwise be prone to escape.


Asunto(s)
Heterocromatina/metabolismo , Elementos de Nucleótido Esparcido Largo , Inactivación del Cromosoma X , Animales , Línea Celular , Células Madre Embrionarias/metabolismo , Femenino , Humanos , Ratones , ARN Largo no Codificante , ARN no Traducido/metabolismo , Transcripción Genética , Cromosoma X/metabolismo
3.
Annu Rev Cell Dev Biol ; 27: 611-29, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21801017

RESUMEN

X-chromosome inactivation, or the silencing of one X chromosome that occurs initially in the female somatic four-cell-stage embryo, is reversed during embryonic development first at the time of inner cell mass formation and again during formation of germ cell precursors. Such X-chromosome reactivation in the mouse implies the silencing of the Xist gene and the transcription of its antisense partner, Tsix, from both X chromosomes. In murine embryonic stem cells, both genes are under the transcriptional control of a series of critical pluripotency factors, namely, OCT3/4, NANOG, SOX2, KLF4, C-MYC and REX1. Although the inactive/active status of the two X chromosomes present in female human embryonic stem cells remains controversial, the reactivation of X-chromosome inactivation seems to be a signature for the naive pluripotent state.


Asunto(s)
Células Madre Pluripotentes/fisiología , Inactivación del Cromosoma X , Cromosoma X/metabolismo , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Epigénesis Genética , Silenciador del Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factor 4 Similar a Kruppel , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , ARN Largo no Codificante , ARN no Traducido/genética , ARN no Traducido/metabolismo , Transducción de Señal/fisiología , Cromosoma X/genética
4.
Mamm Genome ; 28(1-2): 1-12, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27671790

RESUMEN

Nonobese diabetic (NOD) mice are a model for type 1 diabetes that displays defects in central immune tolerance, including impairment of thymocyte apoptosis and proliferation. Thymocyte apoptosis is decreased in NOD/Lt mice compared to nondiabetic C3H/HeJ and C57BL/6 mice. Analysis of a set of NOD.C3H and NOD.B6 congenic mouse strains for distal chromosome 6 localizes the phenotype to the 700 kb Idd6.3 interval. Idd6.3 contains the type 1 diabetes candidate gene aryl hydrocarbon receptor nuclear translocator-like 2 (Arntl2), encoding a circadian rhythm-related transcription factor. Newly generated Arntl2 -/- mouse strains reveal that inactivation of the B6 allele of Arntl2 is sufficient to both decrease thymocyte apoptosis and proliferation. When expressed from C3H or B6 alleles, ARNTL2 inhibits the transcription of interleukin 21 (Il21), a major player in the regulation of immune responses. IL-21 injection abolishes the B6 allele-mediated decrease of apoptosis and proliferation. Interestingly, IL-21 also leads to an increase in thymic proinflammatory Th17 helper cells. Our results identify Arntl2 as a gene controlling thymocyte apoptosis and proliferation along with Th17 development through the IL-21 pathway.


Asunto(s)
Factores de Transcripción ARNTL/genética , Apoptosis/genética , Ritmo Circadiano/genética , Diabetes Mellitus Tipo 1/genética , Animales , Proliferación Celular/genética , Cromosomas/genética , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Ratones , Ratones Endogámicos NOD/genética , Ratones Noqueados , Células Th17/metabolismo , Timocitos/metabolismo , Timocitos/patología
5.
Nature ; 468(7322): 457-60, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21085182

RESUMEN

The reprogramming of X-chromosome inactivation during the acquisition of pluripotency in vivo and in vitro is accompanied by the repression of Xist, the trigger of X-inactivation, and the upregulation of its antisense counterpart Tsix. We have shown that key factors supporting pluripotency-Nanog, Oct4 and Sox2-bind within Xist intron 1 in undifferentiated embryonic stem cells (ESC) to repress Xist transcription. However, the relationship between transcription factors of the pluripotency network and Tsix regulation has remained unclear. Here we show that Tsix upregulation in embryonic stem cells depends on the recruitment of the pluripotent marker Rex1, and of the reprogramming-associated factors Klf4 and c-Myc, by the DXPas34 minisatellite associated with the Tsix promoter. Upon deletion of DXPas34, binding of the three factors is abrogated and the transcriptional machinery is no longer efficiently recruited to the Tsix promoter. Additional analyses including knockdown experiments further demonstrate that Rex1 is critically important for efficient transcription elongation of Tsix. Hence, distinct embryonic-stem-cell-specific complexes couple X-inactivation reprogramming and pluripotency, with Nanog, Oct4 and Sox2 repressing Xist to facilitate the reactivation of the inactive X, and Klf4, c-Myc and Rex1 activating Tsix to remodel Xist chromatin and ensure random X-inactivation upon differentiation. The holistic pattern of Xist/Tsix regulation by pluripotent factors that we have identified suggests a general direct governance of complex epigenetic processes by the machinery dedicated to pluripotency.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes/metabolismo , ARN no Traducido/genética , Transcripción Genética/genética , Regulación hacia Arriba/genética , Animales , Células Madre Embrionarias/citología , Femenino , Proteínas de Homeodominio/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Repeticiones de Minisatélite/genética , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Largo no Codificante , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/metabolismo , Inactivación del Cromosoma X/genética
6.
Stem Cells ; 32(2): 377-90, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24115267

RESUMEN

Random epigenetic silencing of the X-chromosome in somatic tissues of female mammals equalizes the dosage of X-linked genes between the sexes. Unlike this form of X-inactivation that is essentially irreversible, the imprinted inactivation of the paternal X, which characterizes mouse extra-embryonic tissues, appears highly unstable in the trophoblast giant cells of the placenta. Here, we wished to determine whether such instability is already present in placental progenitor cells prior to differentiation toward lineage-specific cell types. To this end, we analyzed the behavior of a GFP transgene on the paternal X both in vivo and in trophoblast stem (TS) cells derived from the trophectoderm of XX(GFP) blastocysts. Using single-cell studies, we show that not only the GFP transgene but also a large number of endogenous genes on the paternal X are subject to orchestrated cycles of reactivation/de novo inactivation in placental progenitor cells. This reversal of silencing is associated with local losses of histone H3 lysine 27 trimethylation extending over several adjacent genes and with the topological relocation of the hypomethylated loci outside of the nuclear compartment of the inactive X. The "reactivated" state is maintained through several cell divisions. Our study suggests that this type of "metastable epigenetic" states may underlie the plasticity of TS cells and predispose specific genes to relaxed regulation in specific subtypes of placental cells.


Asunto(s)
Compensación de Dosificación (Genética) , Genes Ligados a X , Inactivación del Cromosoma X/genética , Cromosoma X/genética , Animales , Blastocisto/metabolismo , Epigénesis Genética , Femenino , Humanos , Ratones , Placenta , Embarazo , Células Madre , Trofoblastos/metabolismo
7.
Nat Rev Immunol ; 3(3): 243-52, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12658272

RESUMEN

Autoimmune diseases are, in general, under complex genetic control and subject to strong interactions between genetics and the environment. Greater knowledge of the underlying genetics will provide immunologists with a framework for study of the immune dysregulation that occurs in such diseases. Ascertaining the number of genes that are involved and their characterization have, however, proven to be difficult. Improved methods of genetic analysis and the availability of a draft sequence of the complete mouse genome have markedly improved the outlook for such research, and they have emphasized the advantages of mice as a model system. In this review, we provide an overview of the genetic analysis of autoimmune diseases and of the crucial role of congenic and consomic mouse strains in such research.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Ratones Congénicos/genética , Ratones Congénicos/inmunología , Animales , Modelos Animales de Enfermedad , Variación Genética/inmunología , Humanos , Ratones , Sitios de Carácter Cuantitativo/inmunología
9.
PLoS Genet ; 7(7): e1002212, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21811421

RESUMEN

In humans, sexual dimorphism is associated with the presence of two X chromosomes in the female, whereas males possess only one X and a small and largely degenerate Y chromosome. How do men cope with having only a single X chromosome given that virtually all other chromosomal monosomies are lethal? Ironically, or even typically many might say, women and more generally female mammals contribute most to the job by shutting down one of their two X chromosomes at random. This phenomenon, called X-inactivation, was originally described some 50 years ago by Mary Lyon and has captivated an increasing number of scientists ever since. The fascination arose in part from the realisation that the inactive X corresponded to a dense heterochromatin mass called the "Barr body" whose number varied with the number of Xs within the nucleus and from the many intellectual questions that this raised: How does the cell count the X chromosomes in the nucleus and inactivate all Xs except one? What kind of molecular mechanisms are able to trigger such a profound, chromosome-wide metamorphosis? When is X-inactivation initiated? How is it transmitted to daughter cells and how is it reset during gametogenesis? This review retraces some of the crucial findings, which have led to our current understanding of a biological process that was initially considered as an exception completely distinct from conventional regulatory systems but is now viewed as a paradigm "par excellence" for epigenetic regulation.


Asunto(s)
Genética/historia , ARN no Traducido/genética , Cromatina Sexual/genética , Inactivación del Cromosoma X , Animales , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Genética/tendencias , Historia del Siglo XX , Historia del Siglo XXI , Humanos , ARN Largo no Codificante
10.
Hum Mol Genet ; 20(4): 705-18, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21118898

RESUMEN

X chromosome inactivation (XCI) is an essential epigenetic process which involves several non-coding RNAs (ncRNAs), including Xist, the master regulator of X-inactivation initiation. Xist is flanked in its 5' region by a large heterochromatic hotspot, which contains several transcription units including a gene of unknown function, Ftx (five prime to Xist). In this article, we describe the characterization and functional analysis of murine Ftx. We present evidence that Ftx produces a conserved functional long ncRNA, and additionally hosts microRNAs (miR) in its introns. Strikingly, Ftx partially escapes X-inactivation and is upregulated specifically in female ES cells at the onset of X-inactivation, an expression profile which closely follows that of Xist. We generated Ftx null ES cells to address the function of this gene. In these cells, only local changes in chromatin marks are detected within the hotspot, indicating that Ftx is not involved in the global maintenance of the heterochromatic structure of this region. The Ftx mutation, however, results in widespread alteration of transcript levels within the X-inactivation center (Xic) and particularly important decreases in Xist RNA levels, which were correlated with increased DNA methylation at the Xist CpG island. Altogether our results indicate that Ftx is a positive regulator of Xist and lead us to propose that Ftx is a novel ncRNA involved in XCI.


Asunto(s)
Cromatina/química , ARN no Traducido/genética , ARN no Traducido/metabolismo , Animales , Bovinos , Línea Celular , Cromatina/metabolismo , Metilación de ADN , Compensación de Dosificación (Genética) , Células Madre Embrionarias/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Mutación , Regiones Promotoras Genéticas , ARN Largo no Codificante , Homología de Secuencia , Regulación hacia Arriba , Inactivación del Cromosoma X/genética
11.
Arch Biochem Biophys ; 534(1-2): 20-6, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23031499

RESUMEN

Neuronal differentiation from neural stem cells into mature neurons is guided by the concerted action of specific transcription factors that stepwise exercise their role in the context of defined chromatin states. Amongst the classes of proteins that influence chromatin compaction and modification are nucleosome assembly proteins (NAPs). Mammals possess several nucleosome assembly protein 1 like proteins (NAP1L) that show either ubiquitous or neuron-restricted expression. The latter group is presumably involved in the process of neuronal differentiation. Mammalian NAP1Ls can potentially form both homo- and hetero-dimers and octamers, in theory allowing thousands of different combinations to be formed. Detailed studies have been performed on several of the NAP1Ls that point to a range of molecular roles, including transcriptional regulation, nuclear import, and control of cell division. This article aims at summarizing current knowledge of the mammalian NAP1L family and its interactions.


Asunto(s)
Neurogénesis , Neuronas/citología , Proteína 1 de Ensamblaje de Nucleosomas/metabolismo , Nucleosomas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Ensamble y Desensamble de Cromatina , Regulación de la Expresión Génica , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Neuronas/metabolismo , Proteína 1 de Ensamblaje de Nucleosomas/genética , Nucleosomas/genética , Unión Proteica , Mapeo de Interacción de Proteínas/métodos , Transcripción Genética
12.
Nat Cell Biol ; 8(3): 293-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16434960

RESUMEN

The initial differential treatment of the two X chromosomes during X-chromosome inactivation is controlled by the X-inactivation centre (Xic). This locus determines how many X chromosomes are present in a cell ('counting') and which X chromosome will be inactivated in female cells ('choice'). Critical control sequences in the Xic include the non-coding RNAs Xist and Tsix, and long-range chromatin elements. However, little is known about the process that ensures that X inactivation is triggered appropriately when more than one Xic is present in a cell. Using three-dimensional fluorescence in situ hybridization (FISH) analysis, we showed that the two Xics transiently colocalize, just before X inactivation, in differentiating female embryonic stem cells. Using Xic transgenes capable of imprinted but not random X inactivation, and Xic deletions that disrupt random X inactivation, we demonstrated that Xic colocalization is linked to Xic function in random X inactivation. Both long-range sequences and the Tsix element, which generates the antisense transcript to Xist, are required for the transient interaction of Xics. We propose that transient colocalization of Xics may be necessary for a cell to determine Xic number and to ensure the correct initiation of X inactivation.


Asunto(s)
Impresión Genómica , ARN no Traducido/fisiología , Células Madre/fisiología , Inactivación del Cromosoma X , Cromosoma X/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Embrión de Mamíferos/citología , Femenino , Hibridación Fluorescente in Situ , Masculino , Ratones , ARN Largo no Codificante , ARN no Traducido/genética
13.
PLoS Biol ; 8(1): e1000276, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20052282

RESUMEN

In placental mammals, inactivation of one of the X chromosomes in female cells ensures sex chromosome dosage compensation. The 17 kb non-coding Xist RNA is crucial to this process and accumulates on the future inactive X chromosome. The most conserved Xist RNA region, the A region, contains eight or nine repeats separated by U-rich spacers. It is implicated in the recruitment of late inactivated X genes to the silencing compartment and likely in the recruitment of complex PRC2. Little is known about the structure of the A region and more generally about Xist RNA structure. Knowledge of its structure is restricted to an NMR study of a single A repeat element. Our study is the first experimental analysis of the structure of the entire A region in solution. By the use of chemical and enzymatic probes and FRET experiments, using oligonucleotides carrying fluorescent dyes, we resolved problems linked to sequence redundancies and established a 2-D structure for the A region that contains two long stem-loop structures each including four repeats. Interactions formed between repeats and between repeats and spacers stabilize these structures. Conservation of the spacer terminal sequences allows formation of such structures in all sequenced Xist RNAs. By combination of RNP affinity chromatography, immunoprecipitation assays, mass spectrometry, and Western blot analysis, we demonstrate that the A region can associate with components of the PRC2 complex in mouse ES cell nuclear extracts. Whilst a single four-repeat motif is able to associate with components of this complex, recruitment of Suz12 is clearly more efficient when the entire A region is present. Our data with their emphasis on the importance of inter-repeat pairing change fundamentally our conception of the 2-D structure of the A region of Xist RNA and support its possible implication in recruitment of the PRC2 complex.


Asunto(s)
ARN no Traducido/genética , Proteínas Represoras/genética , Cromosoma X/genética , Animales , Cromosomas Humanos X/genética , Femenino , Células HeLa , Humanos , Secuencias Repetitivas Esparcidas/genética , Ratones , Conformación de Ácido Nucleico , Filogenia , Proteínas del Grupo Polycomb , ARN Largo no Codificante , Inactivación del Cromosoma X/genética
14.
Curr Res Neurobiol ; 5: 100114, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38020809

RESUMEN

The chromodomain helicase DNA-binding protein 8 (CHD8) is a chromatin remodeler whose mutation is associated, with high penetrance, with autism. Individuals with CHD8 mutations share common symptoms such as autistic behaviour, cognitive impairment, schizophrenia comorbidity, and phenotypic features such as macrocephaly and facial defects. Chd8-deficient mouse models recapitulate most of the phenotypes seen in the brain and other organs of humans. It is known that CHD8 regulates - directly and indirectly - neuronal, autism spectrum disorder (ASDs)-associated genes and long non-coding RNAs (lncRNAs) genes, which, in turn, regulate fundamental aspects of neuronal differentiation and brain development and function. A major characteristic of CHD8 regulation of gene expression is its non-linear and dosage-sensitive nature. CHD8 mutations appear to affect males predominantly, although the reasons for this observed sex bias remain- unknown. We have recently reported that CHD8 directly regulates X chromosome inactivation (XCI) through the transcriptional control of the Xist long non-coding RNA (lncRNA), the master regulator of mammalian XCI. We identified a role for CHD8 in regulating accessibility at the Xist promoter through competitive binding with transcription factors (TFs) at Xist regulatory regions. We speculate here that CHD8 might also regulate accessibility at neuronal/ASD targets through a similar competitive binding mechanism during neurogenesis and brain development. However, whilst such a model can reconcile the phenotypic differences observed in Chd8 knock-down (KD) vs knock-out (KO) mouse models, explaining the observed CHD8 non-linear dosage-dependent activity, it cannot on its own explain the observed disease sex bias.

15.
Bioessays ; 32(7): 581-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20544740

RESUMEN

In mice, dosage compensation of X-linked gene expression is achieved through the inactivation of one of the two X-chromosomes in XX female cells. The complex epigenetic process leading to X-inactivation is largely controlled by Xist and Tsix, two non-coding genes of opposing function. Xist RNA triggers X-inactivation by coating the inactive X, while Tsix is critical for the designation of the active X-chromosome through cis-repression of Xist RNA accumulation. Recently, a plethora of trans-acting factors and cis-regulating elements have been suggested to act as key regulators of either Xist, Tsix or both; these include ubiquitous factors such as Yy1 and Ctcf, developmental proteins such as Nanog, Oct4 and Sox2, and X-linked regulators such as Rnf12. In this paper we summarise recent advances in our knowledge of the regulation of Xist and Tsix in embryonic stem (ES) and differentiating ES cells.


Asunto(s)
Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , ARN no Traducido/genética , Animales , Cromatina/metabolismo , Ratones , ARN Largo no Codificante , ARN no Traducido/metabolismo , Inactivación del Cromosoma X/genética
16.
Nature ; 438(7066): 369-73, 2005 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-16227973

RESUMEN

In mammals, one of the two X chromosomes is inactivated in females to enable dosage compensation for X-linked gene products. In rodents and marsupials, only the X chromosome of paternal origin (Xp) is silenced during early embryogenesis. This could be due to a carry-over effect of the X chromosome's passage through the male germ line, where it becomes transiently silenced together with the Y chromosome, during meiotic sex chromosome inactivation (MSCI). Here we show that Xist (X inactive specific transcript) transgenes, located on autosomes, do not undergo MSCI in the male germ line of mice and yet can induce imprinted cis-inactivation when paternally inherited, with identical kinetics to the Xp chromosome. This suggests that MSCI is not necessary for imprinted X-chromosome inactivation in mice. We also show that the Xp is transcribed, like autosomes, at zygotic gene activation rather than being 'pre-inactivated'. We propose that expression of the paternal Xist gene at zygotic gene activation is sufficient to trigger cis-inactivation of the X chromosome, or of an autosome carrying a Xist transgene.


Asunto(s)
Impresión Genómica/genética , Meiosis/genética , Inactivación del Cromosoma X/genética , Cromosoma X/genética , Animales , Embrión de Mamíferos/metabolismo , Femenino , Silenciador del Gen , Histonas/metabolismo , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Transgénicos , ARN Largo no Codificante , ARN no Traducido/genética , Espermatocitos/metabolismo , Transcripción Genética/genética , Transgenes/genética , Cromosoma Y/genética , Cigoto/metabolismo
17.
Commun Biol ; 4(1): 485, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859315

RESUMEN

Female mammals achieve dosage compensation by inactivating one of their two X chromosomes during development, a process entirely dependent on Xist, an X-linked long non-coding RNA (lncRNA). At the onset of X chromosome inactivation (XCI), Xist is up-regulated and spreads along the future inactive X chromosome. Contextually, it recruits repressive histone and DNA modifiers that transcriptionally silence the X chromosome. Xist regulation is tightly coupled to differentiation and its expression is under the control of both pluripotency and epigenetic factors. Recent evidence has suggested that chromatin remodelers accumulate at the X Inactivation Center (XIC) and here we demonstrate a new role for Chd8 in Xist regulation in differentiating ES cells, linked to its control and prevention of spurious transcription factor interactions occurring within Xist regulatory regions. Our findings have a broader relevance, in the context of complex, developmentally-regulated gene expression.


Asunto(s)
Proteínas de Unión al ADN/genética , Inactivación del Cromosoma X , Cromosoma X/genética , Animales , Proteínas de Unión al ADN/metabolismo , Compensación de Dosificación (Genética) , Femenino , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
18.
Immunogenetics ; 62(9): 585-92, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20676886

RESUMEN

The genetic locus Idd6 is involved in type 1 diabetes development in the non-obese diabetic (NOD) mouse through its effect on the immune system and in particular, on T cell activities. Analysis of congenic strains for Idd6 has established the Aryl hydrocarbon receptor nuclear translocator-like 2 (Arntl2) as a likely candidate gene. In this study we investigate the role of Arntl2 in the autoimmune disease and T cell activation. An Arntl2 expressing plasmid was transfected into CD4(+) T cells by nucleofection. Expression levels of cytokines and CD4(+) T cell activation markers, cell death, apoptosis, and cell proliferation rates were characterized in ex vivo experiments whilst in vivo the transfected cells were transferred into NOD.SCID mice to monitor diabetes development. The results demonstrate that Arntl2 overexpression leads to inhibition of CD4(+) T cell proliferation and decreases in their diabetogenic activity without influence on the expression levels of cytokines, CD4(+) T cell activation markers, cell death, and apoptosis. Our findings suggest that Arntl2 at the Idd6 locus may act via the inhibition of CD4(+) T cell proliferation and the reduction in the diabetogenic activity of CD4(+) T cells to protect against autoimmune type 1 diabetes in the NOD mice.


Asunto(s)
Factores de Transcripción ARNTL/genética , Linfocitos T CD4-Positivos/inmunología , Ritmo Circadiano , Diabetes Mellitus Tipo 1/prevención & control , Activación de Linfocitos , Bazo/inmunología , Regulación hacia Arriba/fisiología , Animales , Apoptosis , Western Blotting , Proliferación Celular , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Plásmidos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/metabolismo , Transfección
19.
Curr Opin Genet Dev ; 16(3): 246-53, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16647851

RESUMEN

The mammalian X-chromosome exists in two flavors, active and inactive, in each cell of the adult female. This phenomenon originates from the process of random choice occurring early in development in a small number of progenitor cells in which the decision is made to inactivate either one or the other X chromosome on a cell-autonomous basis. Once made, this initial decision is irreversible, although exceptions exist in specific chromosomal territories and cell lineages. Recent findings implicate various factors, including non-coding RNAs and chromatin modification complexes, as effectors in the initiation and maintenance of X-chromosome inactivation. The functional redundancy of such factors almost certainly plays an important role in the stability of the inactive X. Studying skewing or bias opens an important opportunity for understanding facets of the random choice process.


Asunto(s)
Inactivación del Cromosoma X/genética , Cromosoma X/genética , Animales , Heterogeneidad Genética , Humanos , Ratones
20.
Nephrol Dial Transplant ; 25(3): 764-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19854849

RESUMEN

BACKGROUND: Female carriers of X-linked Alport syndrome (XLAS) demonstrate variability in clinical phenotype that, unlike males, cannot be correlated with genotype. X-inactivation, the method by which females (XX) silence transcription from one X chromosome in order to achieve gene dosage parity with males (XY), likely modifies the carrier phenotype, but this hypothesis has not been tested directly. METHODS: Using a genetically defined mouse model of XLAS, we generated two groups of Alport female (Col4a5(+/-)) carriers that differed only in the X-controlling element (Xce) allele regulating X-inactivation. We followed the groups as far as 6 months of age comparing survival and surrogate outcome measures of urine protein and plasma urea nitrogen. RESULTS: Preferential inactivation of the mutant Col4a5 gene improved survival and surrogate outcome measures of urine protein and plasma urea nitrogen. In studies of surviving mice, we found that X-inactivation in kidney, measured by allele-specific mRNA expression assays, correlated with surrogate outcomes. CONCLUSIONS: Our findings establish X-inactivation as a major modifier of the carrier phenotype in X-linked Alport syndrome. Thus, X-inactivation patterns may offer prognostic information and point to possible treatment strategies for symptomatic carriers.


Asunto(s)
Modelos Animales de Enfermedad , Nefritis Hereditaria/genética , Índice de Severidad de la Enfermedad , Inactivación del Cromosoma X/genética , Animales , Nitrógeno de la Urea Sanguínea , Colágeno Tipo IV/genética , Femenino , Genotipo , Heterocigoto , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Nefritis Hereditaria/metabolismo , Fenotipo , Proteinuria/orina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA