Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Nat Commun ; 12(1): 3638, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131144

RESUMEN

To ensure dosage compensation between the sexes, one randomly chosen X chromosome is silenced in each female cell in the process of X-chromosome inactivation (XCI). XCI is initiated during early development through upregulation of the long non-coding RNA Xist, which mediates chromosome-wide gene silencing. Cell differentiation, Xist upregulation and gene silencing are thought to be coupled at multiple levels to ensure inactivation of exactly one out of two X chromosomes. Here we perform an integrated analysis of all three processes through allele-specific single-cell RNA-sequencing. Specifically, we assess the onset of random XCI in differentiating mouse embryonic stem cells, and develop dedicated analysis approaches. By exploiting the inter-cellular heterogeneity of XCI onset, we identify putative Xist regulators. Moreover, we show that transient Xist upregulation from both X chromosomes results in biallelic gene silencing right before transitioning to the monoallelic state, confirming a prediction of the stochastic model of XCI. Finally, we show that genetic variation modulates the XCI process at multiple levels, providing a potential explanation for the long-known X-controlling element (Xce) effect, which leads to preferential inactivation of a specific X chromosome in inter-strain crosses. We thus draw a detailed picture of the different levels of regulation that govern the initiation of XCI. The experimental and computational strategies we have developed here will allow us to profile random XCI in more physiological contexts, including primary human cells in vivo.


Asunto(s)
ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba , Inactivación del Cromosoma X , Alelos , Animales , Compensación de Dosificación (Genética) , Femenino , Silenciador del Gen , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones , Análisis de Secuencia de ARN , Cromosoma X , Inactivación del Cromosoma X/genética , Inactivación del Cromosoma X/fisiología
2.
Mol Cell Biol ; 41(8): e0038220, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34060915

RESUMEN

The long noncoding RNA XIST is the master regulator for the process of X chromosome inactivation (XCI) in mammalian females. Here, we report the existence of a hitherto-uncharacterized cis regulatory element (cRE) within the first exon of human XIST, which determines the transcriptional status of XIST during the initiation and maintenance phases of XCI. In the initiation phase, pluripotency factors bind to this cRE and keep XIST repressed. In the maintenance phase of XCI, the cRE is enriched for CTCF, which activates XIST transcription. By employing a CRISPR-dCas9-KRAB-based interference strategy, we demonstrate that binding of CTCF to the newly identified cRE is critical for regulating XIST in a YY1-dependent manner. Collectively, our study uncovers the combinatorial effect of multiple transcriptional regulators influencing XIST expression during the initiation and maintenance phases of XCI.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Células Madre Embrionarias/metabolismo , ARN Largo no Codificante/genética , Inactivación del Cromosoma X/fisiología , Factor de Unión a CCCTC/genética , Humanos , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Inactivación del Cromosoma X/genética
3.
Med Sci (Paris) ; 37(2): 152-158, 2021 Feb.
Artículo en Francés | MEDLINE | ID: mdl-33591258

RESUMEN

The inactivation of one of the two X chromosomes of female mammals is a vital process and a paradigm for epigenetic regulations. X-inactivation is triggered, early during embryo development, by the accumulation of a peculiar noncoding RNA, XIST, which interacts with a plethora of molecular complexes and ultimately protects the coated chromosome from the expression machinery. Once installed, the inactive state is locked by multiple layers of chromatin modifications, ensuring its stable perpetuation across cell divisions. However, recent discoveries made in various model organisms urge us to revisit some of the general principles of the X-inactivation process.


TITLE: Dernières nouvelles du chromosome X - Des principes généraux nuancés. ABSTRACT: L'inactivation d'un des deux chromosomes X des femelles mammifères est un processus vital et emblématique des régulations épigénétiques. Elle est déclenchée par l'accumulation d'un ARN non codant, XIST, qui isole le chromosome concerné de la machinerie transcriptionnelle ; l'état inactif persiste ensuite de manière stable au cours des divisions cellulaires successives. Cependant, des découvertes récentes conduisent à revisiter certains principes généraux de l'inactivation du chromosome X initialement établis. Ainsi le chercheur, tout comme le poète**, est-il invité à « vingt fois sur le métier remettre son ouvrage ¼.


Asunto(s)
Inactivación del Cromosoma X/fisiología , Animales , Femenino , Silenciador del Gen/fisiología , Genética/tendencias , Humanos , Mamíferos/genética , Cromosoma X/genética
4.
Cell Rep ; 33(10): 108485, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33296655

RESUMEN

Immune responses differ between women and men, and type I interferon (IFN) responses following Toll-like receptor 7 (TLR7) stimulation are higher in women. The precise mechanisms driving these sex differences in immunity are unknown. To investigate possible genetic factors, we quantify escape from X chromosome inactivation (XCI) for TLR7 and four other genes (RPS6KA3, CYBB, BTK, and IL13RA1) at the single plasmacytoid dendritic cell (pDC) level. We observe escape from XCI for all investigated genes, leading to biallelic expression patterns. pDCs with biallelic gene expression have significantly higher mRNA levels of the respective genes. Unstimulated pDCs with biallelic TLR7 expression exhibit significantly higher IFNα/ß mRNA levels, and IFNα exposure results in significantly increased IFNα/ß protein production by pDCs. These results identify unanticipated heterogeneity in escape from XCI of several genes in pDCs and highlight the important contribution of X chromosome factors to sex differences in type I IFN responses, which might explain observed sex differences in human diseases.


Asunto(s)
Células Dendríticas/metabolismo , Interferón Tipo I/metabolismo , Receptor Toll-Like 7/metabolismo , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/fisiología , Femenino , Expresión Génica/genética , Humanos , Inmunidad Innata/fisiología , Interferón Tipo I/fisiología , Masculino , ARN Mensajero/metabolismo , Caracteres Sexuales , Factores Sexuales , Transducción de Señal/genética , Inactivación del Cromosoma X/genética , Inactivación del Cromosoma X/fisiología
5.
Nat Cell Biol ; 22(9): 1116-1129, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32807903

RESUMEN

How allelic asymmetry is generated remains a major unsolved problem in epigenetics. Here we model the problem using X-chromosome inactivation by developing "BioRBP", an enzymatic RNA-proteomic method that enables probing of low-abundance interactions and an allelic RNA-depletion and -tagging system. We identify messenger RNA-decapping enzyme 1A (DCP1A) as a key regulator of Tsix, a noncoding RNA implicated in allelic choice through X-chromosome pairing. DCP1A controls Tsix half-life and transcription elongation. Depleting DCP1A causes accumulation of X-X pairs and perturbs the transition to monoallelic Tsix expression required for Xist upregulation. While ablating DCP1A causes hyperpairing, forcing Tsix degradation resolves pairing and enables Xist upregulation. We link pairing to allelic partitioning of CCCTC-binding factor (CTCF) and show that tethering DCP1A to one Tsix allele is sufficient to drive monoallelic Xist expression. Thus, DCP1A flips a bistable switch for the mutually exclusive determination of active and inactive Xs.


Asunto(s)
Endorribonucleasas/metabolismo , ARN/metabolismo , Transactivadores/metabolismo , Cromosoma X/metabolismo , Alelos , Animales , Factor de Unión a CCCTC/metabolismo , Línea Celular , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Transcripción Genética/fisiología , Regulación hacia Arriba/fisiología , Inactivación del Cromosoma X/fisiología
6.
Cells ; 9(4)2020 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-32325818

RESUMEN

X-chromosome inactivation (XCI) is a developmental process that aims to equalize the dosage of X-linked gene products between XY males and XX females in eutherian mammals. In female mouse embryos, paternal XCI is initiated at the 4-cell stage; however, the X chromosome is reactivated in the inner cell mass cells of blastocysts, and random XCI is subsequently initiated in epiblast cells. However, recent findings show that the patterns of XCI are not conserved among mammals. In this study, we used quantitative RT-PCR and RNA in situ hybridization combined with immunofluorescence to investigate the pattern of XCI during bovine embryo development. Expression of XIST (X-inactive specific transcript) RNA was significantly upregulated at the morula stage. For the first time, we demonstrate that XIST accumulation in bovine embryos starts in nuclei of female morulae, but its colocalization with histone H3 lysine 27 trimethylation was first detected in day 7 blastocysts. Both in the inner cell mass and in putative epiblast precursors, we observed a proportion of cells with XIST RNA and H3K27me3 colocalization. Surprisingly, the onset of XCI did not lead to a global downregulation of X-linked genes, even in day 9 blastocysts. Together, our findings confirm that diverse patterns of XCI initiation exist among developing mammalian embryos.


Asunto(s)
Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Inactivación del Cromosoma X/fisiología , Animales , Blastocisto/metabolismo , Bovinos , Metilación de ADN , Impresión Genómica/genética , Histonas/metabolismo , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
7.
Nat Commun ; 11(1): 764, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32034154

RESUMEN

Our understanding of the signalling pathways regulating early human development is limited, despite their fundamental biological importance. Here, we mine transcriptomics datasets to investigate signalling in the human embryo and identify expression for the insulin and insulin growth factor 1 (IGF1) receptors, along with IGF1 ligand. Consequently, we generate a minimal chemically-defined culture medium in which IGF1 together with Activin maintain self-renewal in the absence of fibroblast growth factor (FGF) signalling. Under these conditions, we derive several pluripotent stem cell lines that express pluripotency-associated genes, retain high viability and a normal karyotype, and can be genetically modified or differentiated into multiple cell lineages. We also identify active phosphoinositide 3-kinase (PI3K)/AKT/mTOR signalling in early human embryos, and in both primed and naïve pluripotent culture conditions. This demonstrates that signalling insights from human blastocysts can be used to define culture conditions that more closely recapitulate the embryonic niche.


Asunto(s)
Autorrenovación de las Células/fisiología , Células Madre Embrionarias Humanas/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Activinas/metabolismo , Animales , Blastocisto/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo/química , Medios de Cultivo/metabolismo , Medios de Cultivo/farmacología , Endodermo/citología , Endodermo/metabolismo , Membranas Extraembrionarias/citología , Membranas Extraembrionarias/metabolismo , Fibroblastos/citología , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/fisiología , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Transcriptoma , Inactivación del Cromosoma X/fisiología
8.
FASEB J ; 34(1): 691-705, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914626

RESUMEN

The inner cell mass (ICM) in blastocyst is the origin of all somatic and germ cells in mammals and pluripotent stem cells (PSCs) in vitro. As the conserved principles between pig and human, here we performed comprehensive single-cell RNA-seq for porcine early embryos from oocyte to early blastocyst (EB). We show the specification of the ICM and trophectoderm in morula and the molecular signature of the precursors. We demonstrate the existence of naïve pluripotency signature in morula and ICM of EB, and the specific pluripotent genes and the activity of signalling pathways highlight the characteristics of the naïve pluripotency. We observe the absence of dosage compensation with respect to X-chromosome (XC) in morula, and incomplete dosage compensation in the EB. However, the dynamics of dosage compensation may be independent of the expression of XIST induced XC inactivation. Our study describes molecular landmarks of embryogenesis in pig that will provide a better strategy for derivation of porcine PSCs and improve research in regenerative medicine.


Asunto(s)
Blastocisto/citología , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica/genética , Estratos Germinativos/citología , Oocitos/citología , Animales , Perfilación de la Expresión Génica/métodos , Células Germinativas/citología , Células Madre Pluripotentes/citología , Porcinos , Inactivación del Cromosoma X/fisiología
9.
Essays Biochem ; 63(6): 663-676, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31782494

RESUMEN

Monoallelic gene expression occurs in diploid cells when only one of the two alleles of a gene is active. There are three main classes of genes that display monoallelic expression in mammalian genomes: (1) imprinted genes that are monoallelically expressed in a parent-of-origin dependent manner; (2) X-linked genes that undergo random X-chromosome inactivation in female cells; (3) random monoallelically expressed single and clustered genes located on autosomes. The heritability of monoallelic expression patterns during cell divisions implies that epigenetic mechanisms are involved in the cellular memory of these expression states. Among these, methylation of CpG sites on DNA is one of the best described modification to explain somatic inheritance. Here, we discuss the relevance of DNA methylation for the establishment and maintenance of monoallelic expression patterns among these three groups of genes, and how this is intrinsically linked to development and cellular states.


Asunto(s)
Alelos , Metilación de ADN , ADN/metabolismo , Epigénesis Genética/fisiología , Expresión Génica/fisiología , Animales , ADN/genética , Metilación de ADN/fisiología , Femenino , Impresión Genómica/fisiología , Humanos , Inactivación del Cromosoma X/fisiología
10.
J Reprod Dev ; 65(6): 533-539, 2019 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-31631092

RESUMEN

Xist is an X-linked ribonucleic acid (RNA) gene responsible for the cis induction of X chromosome inactivation (XCI). In cloned mammalian embryos, Xist is ectopically activated at the morula to blastocyst stage on the X chromosome that is supposed to be active, thus resulting in abnormal XCI. Suppression of erroneous Xist expression by injecting small interfering RNA (siRNA) remarkably increased the developmental efficiency of cloned male mouse embryos by approximately 10-fold. However, injection of anti-Xist siRNA resulted in only a slight increase in the developmental ability of injected cloned male pig embryos because the blocking effect of the injected siRNA was not maintained beyond the morula stage, which is 5 days post-activation. To develop a more effective approach for suppressing the ectopic expression of Xist in cloned pig embryos, we compared the silencing effect of short hairpin RNA (shRNA) and siRNA on Xist expression and the effects of these two Xist knockdown methods on the developmental competence of cloned male pig embryos. Results indicated that an shRNA-based RNA interference (RNAi) has a longer blocking effect on Xist expression than an siRNA-mediated RNAi. Injection of anti-Xist shRNA plasmid into two-cell-stage cloned male pig embryos effectively suppressed Xist expression, rescued XCI at the blastocyst stage, and improved the in vitro developmental ability of injected cloned embryos. These positive effects, however, were not observed in cloned male pig embryos injected with anti-Xist siRNA. This study demonstrates that vector-based rather than siRNA-mediated RNAi of Xist expression can be employed to improve pig cloning efficiency.


Asunto(s)
Clonación de Organismos/métodos , Desarrollo Embrionario , Interferencia de ARN/fisiología , ARN Largo no Codificante/genética , ARN Interferente Pequeño/genética , Inactivación del Cromosoma X/fisiología , Animales , Animales Modificados Genéticamente , Células Cultivadas , Embrión de Mamíferos , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Técnicas de Silenciamiento del Gen/veterinaria , Vectores Genéticos , Masculino , Técnicas de Transferencia Nuclear , ARN Interferente Pequeño/farmacología , Porcinos/embriología , Porcinos/genética
11.
Artículo en Inglés | MEDLINE | ID: mdl-31450876

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder with a genetic basis that is associated with the mutation of the X-linked methyl-CpG binding protein 2 (MECP2) gene in approximately 90% of patients. RTT is characterized by a brief period of normal development followed by loss of acquired skills and evolution towards impairment of brain and motor functions and multi-organ dysfunction. Originally, RTT was considered lethal in males as it has an X-linked dominant inheritance. However, although this syndrome has a higher incidence in females, rare cases are also documented in males. Here, we describe the case of an 11-year-old male patient with a microduplication MECP2 Xq28. Our patient is currently living, while his older brother with the same mutation died at the age of 9 years. We showed that the role of MECP2 as an epigenetic modulator and the X-chromosome inactivation pattern can explain the lethal clinical form of the older brother with the same microduplication MECP2 Xq28 presented by our patient who is still alive. Given the limited case history of RTT in males, further studies are needed to better characterize this syndrome in males and consequently improve the currently available therapeutic strategies.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/genética , Síndrome de Rett/genética , Inactivación del Cromosoma X/genética , Niño , Compensación de Dosificación (Genética) , Humanos , Masculino , Proteína 2 de Unión a Metil-CpG/fisiología , Mutación , Evaluación del Resultado de la Atención al Paciente , Hermanos , Inactivación del Cromosoma X/fisiología
13.
EMBO J ; 38(12)2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31088843

RESUMEN

Human pluripotent stem cells (hPSCs) are being increasingly utilized worldwide in investigating human development, and modeling and discovering therapies for a wide range of diseases as well as a source for cellular therapy. Yet, since the first isolation of human embryonic stem cells (hESCs) 20 years ago, followed by the successful reprogramming of human-induced pluripotent stem cells (hiPSCs) 10 years later, various studies shed light on abnormalities that sometimes accumulate in these cells in vitro Whereas genetic aberrations are well documented, epigenetic alterations are not as thoroughly discussed. In this review, we highlight frequent epigenetic aberrations found in hPSCs, including alterations in DNA methylation patterns, parental imprinting, and X chromosome inactivation. We discuss the potential origins of these abnormalities in hESCs and hiPSCs, survey the different methods for detecting them, and elaborate on their potential consequences for the different utilities of hPSCs.


Asunto(s)
Epigénesis Genética/fisiología , Células Madre Pluripotentes/fisiología , Diferenciación Celular/genética , Reprogramación Celular/genética , Metilación de ADN/fisiología , Impresión Genómica/genética , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Inactivación del Cromosoma X/fisiología
14.
Prenat Diagn ; 39(8): 603-608, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31069818

RESUMEN

OBJECTIVE: To predict the risk of dystrophinopathy in fetal carriers of dystrophin gene (DMD) mutations. METHODS: Twenty-three pregnant women, with a total of 25 female fetuses carrying DMD mutations, were recruited. Among them, 13 pregnant women who participated in this study were only used to analyse the incidence of induced abortion after fetuses were diagnosed as dystrophinopathy carriers. Eleven fetal carriers from 10 pregnant women were tested to analyse X-chromosome inactivation (XCI) using amniocytes to assess the risk of dystrophinopathy. Follow-ups were conducted on all cases. RESULTS: Approximately one-third of fetuses were aborted before assessing the risk of dystrophinopathy. XCI analysis of amniocytes showed that 10 fetuses had random XCI patterns, and one fetus exhibited a highly skewed XCI pattern (100:0) with primary expression of the maternal X chromosome that carried the mutant allele. These 11 fetal carriers were born, and follow-up showed that the girl who showed the skewed XCI pattern as a fetus was diagnosed with Duchenne muscular dystrophy (DMD) at the age of four. The others did not present with dystrophinopathy-associated symptoms. CONCLUSIONS: XCI was significantly implicated in symptomatic female carriers of dystrophinopathies, and XCI pattern analysis of amniocytes may be useful in predicting the risk of dystrophinopathy in fetal carriers.


Asunto(s)
Amnios/metabolismo , Distrofina/genética , Feto/metabolismo , Distrofia Muscular de Duchenne/diagnóstico , Inactivación del Cromosoma X/fisiología , Aborto Inducido/estadística & datos numéricos , Adulto , Amnios/patología , Estudios de Cohortes , Femenino , Pruebas Genéticas , Heterocigoto , Humanos , Incidencia , Recién Nacido , Masculino , Distrofia Muscular de Duchenne/epidemiología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Mutación , Linaje , Fenotipo , Embarazo , Diagnóstico Prenatal/métodos , Pronóstico , Factores de Riesgo
15.
Sci Rep ; 9(1): 6068, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30988473

RESUMEN

X chromosome inactivation (XCI) is a mammalian specific, developmentally regulated process relying on several mechanisms including antisense transcription, non-coding RNA-mediated silencing, and recruitment of chromatin remodeling complexes. In vitro modeling of XCI, through differentiation of embryonic stem cells (ESCs), provides a powerful tool to study the dynamics of XCI, overcoming the need for embryos, and facilitating genetic modification of key regulatory players. However, to date, robust initiation of XCI in vitro has been mostly limited to mouse pluripotent stem cells. Here, we adapted existing protocols to establish a novel monolayer differentiation protocol for rat ESCs to study XCI. We show that differentiating rat ESCs properly downregulate pluripotency factor genes, and present female specific Xist RNA accumulation and silencing of X-linked genes. We also demonstrate that RNF12 seems to be an important player in regulation of initiation of XCI in rat, acting as an Xist activator. Our work provides the basis to investigate the mechanisms directing the XCI process in a model organism different from the mouse.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/fisiología , ARN Largo no Codificante/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Inactivación del Cromosoma X/fisiología , Animales , Células Cultivadas , Embrión de Mamíferos , Femenino , Masculino , Modelos Animales , Cultivo Primario de Células , Ratas
16.
Nat Commun ; 10(1): 500, 2019 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-30700715

RESUMEN

High-resolution molecular programmes delineating the cellular foundations of mammalian embryogenesis have emerged recently. Similar analysis of human embryos is limited to pre-implantation stages, since early post-implantation embryos are largely inaccessible. Notwithstanding, we previously suggested conserved principles of pig and human early development. For further insight on pluripotent states and lineage delineation, we analysed pig embryos at single cell resolution. Here we show progressive segregation of inner cell mass and trophectoderm in early blastocysts, and of epiblast and hypoblast in late blastocysts. We show that following an emergent short naive pluripotent signature in early embryos, there is a protracted appearance of a primed signature in advanced embryonic stages. Dosage compensation with respect to the X-chromosome in females is attained via X-inactivation in late epiblasts. Detailed human-pig comparison is a basis towards comprehending early human development and a foundation for further studies of human pluripotent stem cell differentiation in pig interspecies chimeras.


Asunto(s)
Análisis de la Célula Individual/métodos , Cromosoma X/metabolismo , Animales , Diferenciación Celular/fisiología , Femenino , Gastrulación/fisiología , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/metabolismo , Humanos , Porcinos , Inactivación del Cromosoma X/fisiología
17.
Cell ; 176(1-2): 182-197.e23, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30595450

RESUMEN

During development, the precise relationships between transcription and chromatin modifications often remain unclear. We use the X chromosome inactivation (XCI) paradigm to explore the implication of chromatin changes in gene silencing. Using female mouse embryonic stem cells, we initiate XCI by inducing Xist and then monitor the temporal changes in transcription and chromatin by allele-specific profiling. This reveals histone deacetylation and H2AK119 ubiquitination as the earliest chromatin alterations during XCI. We show that HDAC3 is pre-bound on the X chromosome and that, upon Xist coating, its activity is required for efficient gene silencing. We also reveal that first PRC1-associated H2AK119Ub and then PRC2-associated H3K27me3 accumulate initially at large intergenic domains that can then spread into genes only in the context of histone deacetylation and gene silencing. Our results reveal the hierarchy of chromatin events during the initiation of XCI and identify key roles for chromatin in the early steps of transcriptional silencing.


Asunto(s)
Cromatina/metabolismo , Inactivación del Cromosoma X/genética , Inactivación del Cromosoma X/fisiología , Acetilación , Animales , Cromatina/genética , Células Madre Embrionarias , Epigenómica/métodos , Femenino , Silenciador del Gen , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Ratones , Proteínas del Grupo Polycomb/metabolismo , Procesamiento Proteico-Postraduccional , ARN Largo no Codificante/metabolismo , Transcripción Genética , Ubiquitinación , Cromosoma X/metabolismo
18.
Am J Med Genet B Neuropsychiatr Genet ; 180(6): 415-427, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30537437

RESUMEN

Co-morbid chronic musculoskeletal pain (CMSP) and posttraumatic stress symptoms (PTSS) are frequent sequelae of motor vehicle collision, are associated with greater disability than either outcome alone, and are more prevalent in women than men. In the current study we assessed for evidence that gene transcripts originating from the X chromosome contribute to sex differences in vulnerability to CMSP and PTSS after motor vehicle collision. Nested samples were drawn from a longitudinal study of African American individuals, and CMSP (0-10 numeric rating scale) and PTSS (impact of events scale, revised) outcomes were assessed 6 months following motor vehicle collision. Blood RNA were sequenced (n = 101) and the relationship between X chromosome mRNA expression levels and co-morbid CMSP and PTSS outcomes was evaluated using logistic regression analyses. A disproportionate number of peritraumatic X chromosome mRNA predicting CMSP and PTSS in women were genes previously found to escape X chromosome inactivation (11/40, z = -2.9, p = .004). Secondary analyses assessing gene ontology relationships between these genes identified an enrichment in genes known to influence neuronal plasticity. Further, the relationship of expression of two critical regulators of X chromosome inactivation, X-inactive specific transcript (XIST) and Yin Yang 1 (YY1), was different in women developing CMSP and PTSS. Together, these data suggest that X chromosome genes that escape inactivation may contribute to sex differences in vulnerability to CMSP and PTSS after motor vehicle collision.


Asunto(s)
Dolor Musculoesquelético/genética , Trastornos por Estrés Postraumático/genética , Inactivación del Cromosoma X/genética , Accidentes de Tránsito/psicología , Adulto , Negro o Afroamericano , Cromosomas Humanos X/genética , Cromosomas Humanos X/fisiología , Comorbilidad , Femenino , Regulación de la Expresión Génica/genética , Humanos , Estudios Longitudinales , Persona de Mediana Edad , Prevalencia , Inactivación del Cromosoma X/fisiología
19.
Hum Mol Genet ; 28(8): 1331-1342, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30576442

RESUMEN

X chromosome inactivation (XCI) is a key epigenetic gene expression regulatory process, which may play a role in women's cancer. In particular tissues, some genes are known to escape XCI, yet patterns of XCI in ovarian cancer (OC) and their clinical associations are largely unknown. To examine XCI in OC, we integrated germline genotype with tumor copy number, gene expression and DNA methylation information from 99 OC patients. Approximately 10% of genes showed different XCI status (either escaping or being subject to XCI) compared with the studies of other tissues. Many of these genes are known oncogenes or tumor suppressors (e.g. DDX3X, TRAPPC2 and TCEANC). We also observed strong association between cis promoter DNA methylation and allele-specific expression imbalance (P = 2.0 × 10-10). Cluster analyses of the integrated data identified two molecular subgroups of OC patients representing those with regulated (N = 47) and dysregulated (N = 52) XCI. This XCI cluster membership was associated with expression of X inactive specific transcript (P = 0.002), a known driver of XCI, as well as age, grade, stage, tumor histology and extent of residual disease following surgical debulking. Patients with dysregulated XCI (N = 52) had shorter time to recurrence (HR = 2.34, P = 0.001) and overall survival time (HR = 1.87, P = 0.02) than those with regulated XCI, although results were attenuated after covariate adjustment. Similar findings were observed when restricted to high-grade serous tumors. We found evidence of a unique OC XCI profile, suggesting that XCI may play an important role in OC biology. Additional studies to examine somatic changes with paired tumor-normal tissue are needed.


Asunto(s)
Carcinoma Epitelial de Ovario/genética , Genes Ligados a X/genética , Inactivación del Cromosoma X/fisiología , Anciano , Alelos , Carcinoma Epitelial de Ovario/metabolismo , Cromosomas Humanos X/genética , Análisis por Conglomerados , Metilación de ADN/genética , Epigénesis Genética/genética , Femenino , Regulación de la Expresión Génica/genética , Frecuencia de los Genes/genética , Estudios de Asociación Genética/métodos , Genotipo , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/genética , Regiones Promotoras Genéticas/genética , ARN Largo no Codificante , Factores de Transcripción/genética , Inactivación del Cromosoma X/genética
20.
Chromosome Res ; 26(3): 179-189, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29679205

RESUMEN

In female mammals, each cell silences one X chromosome by converting it into transcriptionally inert heterochromatin. The inactivation is concomitant with epigenetic changes including methylation of specific histone residues and incorporation of macroH2A. Such epigenetic changes may exert influence on the positioning of the inactive X chromosome (Xi) within the nucleus beyond the level of chromatin structure. However, the dynamic positioning of the inactive X chromosome during cell cycle remains unclear. Here, we show that H3K27me3 is a cell-cycle-independent marker for the inactivated X chromosomes in WI38 cells. By utilizing this marker, three types of Xi locations in the nuclei are classified, which are envelope position (associated with envelope), mid-position (between the envelope and nucleolus), and nucleolus position (associated with the nucleolus). Moreover, serial-section analysis revealed that the inactive X chromosomes in the mid-position appear to be sparser and less condensed than those associated with the nuclear envelope or nucleolus. During the transition from G0 to G1 phase, the inactive X chromosomes tend to move from the envelope position to the nucleolus position in WI38 cells. Our results imply a role of chromosome positioning in maintaining the organization of the inactive X chromosomes in different cell phases.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Fase G1/fisiología , Fase de Descanso del Ciclo Celular/fisiología , Inactivación del Cromosoma X/fisiología , Cromosoma X/metabolismo , Animales , Línea Celular , Femenino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA