Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Vis Exp ; (147)2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31180354

RESUMEN

X chromosome inactivation (XCI) is the random silencing of one X chromosome in females to achieve gene dosage balance between the sexes. As a result, all females are heterozygous for X-linked gene expression. One of the key regulators of XCI is Xist, which is essential for the initiation and maintenance of XCI. Previous studies have identified 13 trans acting X chromosome inactivation factors (XCIFs) using a large-scale, loss-of-function genetic screen. Inhibition of XCIFs, such as ACVR1 and PDPK1, using short-hairpin RNA or small molecule inhibitors, reactivates X chromosome-linked genes in cultured cells. But the feasibility and tolerability of reactivating the inactive X chromosome in vivo remains to be determined. Towards this goal, a XistΔ:Mecp2/Xist:Mecp2-Gfp mouse model has been generated with non-random XCI due to deletion of Xist on one X chromosome. Using this model, the extent of inactive X reactivation was quantitated in the mouse brain following treatment with XCIF inhibitors. Recently published results show, for the first time, that pharmacological inhibition of XCIFs reactivates Mecp2 from the inactive X chromosome in cortical neurons of the living mouse brain.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/genética , Modelos Biológicos , Inactivación del Cromosoma X/genética , Animales , Femenino , Ratones , Ratones Noqueados , Neuronas/metabolismo , ARN Largo no Codificante/genética , Eliminación de Secuencia , Bibliotecas de Moléculas Pequeñas/farmacología , Cromosoma X/genética , Inactivación del Cromosoma X/efectos de los fármacos
2.
ACS Nano ; 13(2): 2050-2061, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30650303

RESUMEN

The widespread use of silver nanoparticles (AgNPs) has raised substantial health risks to human beings. Despite a wealth of progress on toxicity studies, the understanding of the adverse effects on fetuses, embryos, and early stage cells is still rather limited, particularly under low-dose exposure settings. Moreover, nearly all previous studies ascribed AgNP-induced toxic effects to oxidative stress. Differently, we here unearthed a mechanism, namely, interruption of X chromosome inactivation (XCI) in female mouse embryonic stem cells (mESCs). Albeit with no observable cytotoxicity, significant differentiation retardation was found in female mESCs upon low-dose AgNP exposure. Mechanistic investigations uncovered expedited inactivation for the inactive X chromosome (Xi) and attenuated maintenance of the active X chromosome (Xa) state during mESC differentiation upon the challenge of low-dose AgNPs, indicative of disordered XCI. Thereby, a few X-linked genes (which are closely involved in orchestrating ESC differentiation) were found to be repressed, partially attributable to reinforced enrichment of histone modification ( e. g., histone 3 lysine 27 trimethylation, H3K27me3) on their promoter regions, as the result of disordered XCI. In stark contrast to female mESCs, no impairment of differentiation was observed in male mESCs under low-dose AgNP exposure. All considered, our data unearthed that AgNPs at low concentrations compromised the differentiation program of female mESCs through disturbing XCI. Thus, this work would provide a model for the type of studies necessary to advance the understandings on AgNP-induced developmental toxicity.


Asunto(s)
Células Madre Embrionarias/efectos de los fármacos , Nanopartículas del Metal/química , Plata/farmacología , Inactivación del Cromosoma X/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Ratones , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Plata/química , Propiedades de Superficie
3.
Theriogenology ; 110: 79-85, 2018 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-29353144

RESUMEN

This study examines the effects of the histone deacetylation inhibitor scriptaid (SCR) on preimplantation embryo development in vitro and on imprinting gene expression. We hypothesized that SCR would increase histone acetylation levels, enhance embryonic genome activation, and regulate imprinting and X-chromosome inactivation (XCI) in in vitro produced bovine embryos. Zygotes were cultured in vitro in presence or absence of SCR added at different time points. We assessed cleavage and blastocyst rates as well as the quality of blastocysts through: (i) differential cell counts; (ii) survival after vitrification/thawing and (iii) gene expression analysis -including imprinted genes. Blastocyst yields were not different in the control and experimental groups. While no significant differences were observed between groups in total cell or trophectoderm cell numbers, SCR treatment reduced the number of inner cell mass cells and improved the survival of vitrified embryos. Further, genes involved in the mechanism of paternal imprinting (GRB10, GNAS, XIST) were downregulated in presence of SCR compared with controls. These observations suggest SCR prevents deacetylation of paternally imprinting control regions and/or their up-regulation, as these events took place in controls. Whether or not such reductions in XIST and imprinting gene expression are beneficial for post implantation development remains to be clarified.


Asunto(s)
Bovinos/embriología , Desarrollo Embrionario/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Impresión Genómica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Hidroxilaminas/farmacología , Quinolinas/farmacología , Animales , Células Cultivadas , Técnicas de Cultivo de Embriones , Embrión de Mamíferos , Femenino , Embarazo , Inactivación del Cromosoma X/efectos de los fármacos
4.
Proc Natl Acad Sci U S A ; 113(50): 14366-14371, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-28182563

RESUMEN

X-chromosome inactivation is a mechanism of dosage compensation in which one of the two X chromosomes in female mammals is transcriptionally silenced. Once established, silencing of the inactive X (Xi) is robust and difficult to reverse pharmacologically. However, the Xi is a reservoir of >1,000 functional genes that could be potentially tapped to treat X-linked disease. To identify compounds that could reactivate the Xi, here we screened ∼367,000 small molecules in an automated high-content screen using an Xi-linked GFP reporter in mouse fibroblasts. Given the robust nature of silencing, we sensitized the screen by "priming" cells with the DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (5azadC). Compounds that elicited GFP activity include VX680, MLN8237, and 5azadC, which are known to target the Aurora kinase and DNA methylation pathways. We demonstrate that the combinations of VX680 and 5azadC, as well as MLN8237 and 5azadC, synergistically up-regulate genes on the Xi. Thus, our work identifies a synergism between the DNA methylation and Aurora kinase pathways as being one of interest for possible pharmacological reactivation of the Xi.


Asunto(s)
Aurora Quinasas/antagonistas & inhibidores , Metilación de ADN/efectos de los fármacos , Inactivación del Cromosoma X/efectos de los fármacos , Animales , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/genética , Aurora Quinasa B/antagonistas & inhibidores , Aurora Quinasa B/genética , Aurora Quinasas/genética , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Azepinas/administración & dosificación , Línea Celular , Decitabina , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Femenino , Técnicas de Silenciamiento del Gen , Genes Ligados a X , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ensayos Analíticos de Alto Rendimiento , Ratones , Ratones Transgénicos , Piperazinas/administración & dosificación , Pirimidinas/administración & dosificación , Cromosoma X/efectos de los fármacos , Cromosoma X/genética
5.
Epigenetics ; 10(9): 810-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26178744

RESUMEN

In many whole genome studies of gene expression or modified cytosines, data from probes localized to the X-chromosome are removed from analyses due to gender bias. Previously, we observed population differences in cytosine modifications between Caucasian and African lymphoblastoid cell lines (LCLs) on the autosomes using whole genome arrays to measure modified cytosines. DNA methylation plays a critical role in establishment and maintenance of X-chromosome inactivation in females. Therefore, we reasoned that by investigating cytosine modification patterns specifically on the X-chromosome, we could obtain valuable information about a chromosome that is often disregarded in genome-wide analyses. We investigated population differences in cytosine modification patterns along the X-chromosome between Caucasian and African LCLs and identified novel sites that escape methylation on the inactive X-chromosome (Xi) in females. We characterized the chromatin state of these loci by incorporating the extensive histone modification ChIP-seq data generated by ENCODE. To explore the relationship between DNA and histone modifications further, we hypothesized that BRD4, a protein that binds acetylated histones, could be preventing some sites from becoming de novo methylated. To test this, we treated 4 female LCLs with JQ1, a small molecule inhibitor of BRD4, but found that JQ1 treatment induced minor changes in cytosine modification levels, and the majority of sites escaping methylation on the Xi remained unmethylated. This suggests that other epigenetic mechanisms or transcription factors are likely playing a larger role in protecting these sites from de novo methylation on the Xi.


Asunto(s)
Población Negra/genética , Cromosomas Humanos X/metabolismo , Islas de CpG , Metilación de ADN , Población Blanca/genética , Inactivación del Cromosoma X , Azepinas/farmacología , Proteínas de Ciclo Celular , Línea Celular , Cromatina/metabolismo , Cromosomas Humanos X/efectos de los fármacos , Islas de CpG/efectos de los fármacos , Citosina/metabolismo , Metilación de ADN/efectos de los fármacos , Femenino , Humanos , Masculino , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Triazoles/farmacología , Inactivación del Cromosoma X/efectos de los fármacos
6.
Proc Natl Acad Sci U S A ; 111(35): 12591-8, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25136103

RESUMEN

X-chromosome inactivation (XCI), the random transcriptional silencing of one X chromosome in somatic cells of female mammals, is a mechanism that ensures equal expression of X-linked genes in both sexes. XCI is initiated in cis by the noncoding Xist RNA, which coats the inactive X chromosome (Xi) from which it is produced. However, trans-acting factors that mediate XCI remain largely unknown. Here, we perform a large-scale RNA interference screen to identify trans-acting XCI factors (XCIFs) that comprise regulators of cell signaling and transcription, including the DNA methyltransferase, DNMT1. The expression pattern of the XCIFs explains the selective onset of XCI following differentiation. The XCIFs function, at least in part, by promoting expression and/or localization of Xist to the Xi. Surprisingly, we find that DNMT1, which is generally a transcriptional repressor, is an activator of Xist transcription. Small-molecule inhibitors of two of the XCIFs can reversibly reactivate the Xi, which has implications for treatment of Rett syndrome and other dominant X-linked diseases. A homozygous mouse knockout of one of the XCIFs, stanniocalcin 1 (STC1), has an expected XCI defect but surprisingly is phenotypically normal. Remarkably, X-linked genes are not overexpressed in female Stc1(-/-) mice, revealing the existence of a mechanism(s) that can compensate for a persistent XCI deficiency to regulate X-linked gene expression.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Glicoproteínas/genética , Proteína 2 de Unión a Metil-CpG/genética , ARN Largo no Codificante/genética , Síndrome de Rett/genética , Inactivación del Cromosoma X/genética , Animales , Corteza Cerebral/citología , Cromonas/farmacología , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Células Madre Embrionarias/fisiología , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/genética , Femenino , Fibroblastos/citología , Fibroblastos/fisiología , Biblioteca de Genes , Terapia Genética/métodos , Humanos , Mamíferos , Ratones , Ratones Noqueados , Morfolinas/farmacología , Neuronas/citología , Neuronas/fisiología , Pirazoles/farmacología , ARN Interferente Pequeño/genética , Síndrome de Rett/terapia , Sulfonamidas/farmacología , Transcriptoma , Inactivación del Cromosoma X/efectos de los fármacos
7.
Stem Cells ; 32(10): 2642-56, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24965076

RESUMEN

Although the reactivation of silenced X chromosomes has been observed as part of the process of reprogramming female somatic cells into induced pluripotent stem cells (iPSCs), it remains unknown whether repression of the X-inactive specific transcript (Xist) can greatly enhance female iPSC induction similar to that observed in somatic cell nuclear transfer studies. In this study, we discovered that the repression of Xist plays opposite roles in the early and late phases of female iPSCs induction. Our results demonstrate that the downregulation of Xist by an isopropyl ß-d-1-thiogalactopyranoside (IPTG)-inducible short hairpin RNA (shRNA) system can greatly impair the mesenchymal-to-epithelial transition (MET) in the early phase of iPSC induction but can significantly promote the transition of pre-iPSCs to iPSCs in the late phase. Furthermore, we demonstrate that although the knockdown of Xist did not affect the H3K27me3 modification on the X chromosome, macroH2A was released from the inactivated X chromosome (Xi). This enables the X chromosome silencing to be a reversible event. Moreover, we demonstrate that the supplementation of vitamin C (Vc) can augment and stabilize the reversible X chromosome by preventing the relocalization of macroH2A to the Xi. Therefore, our study reveals an opposite role of Xist repression in the early and late stages of reprogramming female somatic cells to pluripotency and demonstrates that the release of macroH2A by Xist repression enables the transition from pre-iPSCs to iPSCs.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Ácido Ascórbico/farmacología , Reprogramación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Masculino , Ratones , Factores de Tiempo , Cromosoma X/genética , Inactivación del Cromosoma X/efectos de los fármacos
8.
J Toxicol Sci ; 38(3): 485-94, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23719926

RESUMEN

Recent studies have reported that bisphenol A (BPA) influences brain development in fetal exposure to mice. The X-chromosome codes many neurodevelopment-related genes leading to abnormal development, such as mental retardation and intellectual deficiency. For females, most of expressions of X-linked genes are regulated by X-chromosome inactivation (XCI), which occurs during fetal period, and this mechanism is regulated by Xist and its antisense, Tsix. To clarify the possibility of X-mediated effect as a mechanism of neurodevelopmental disorders by BPA, pregnant ICR mice were orally administered 0.02 or 50 mg/kg of BPA on gestational days 6 and 15. Postnatally at days 2, 4 and weeks 3 and 7, mRNA expression of XCI-regulating factors (Xist and Tsix), X-linked neurodevelopment-related genes (Fmr1, Gdi1, Nlgn3, Pak3 and Ophn1), and sexual differentiation-related genes (ERα, ERß and AR) were examined in cerebrums of female pups. Anogenital distance (AGD) and serum estradiol were also examined. In the 50 mg/kg exposed-group, reduced Xist, Fmr1, Gdi1, Nlgn3, and Pak3 and increased Tsix were observed simultaneously. Moderately reduced Xist, Gdi1, Nlgn3 and Pak3 were observed at 0.02 mg/kg BPA. ERα, ERß and AR expression changes, shortened AGDs and reduced estradiol levels were observed in each exposure group. Fetal exposure to BPA changed expression of XCI-regulating factors and may alter the expression levels of X-linked neurodevelopment-related genes disrupting the XCI mechanism and function. This X-mediated effect is considered one of the mechanisms of various BPA-induced neurodevelopmental disorders.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Encéfalo/anomalías , Encéfalo/embriología , Estrógenos no Esteroides/toxicidad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Exposición Materna/efectos adversos , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal/genética , ARN Largo no Codificante/fisiología , Inactivación del Cromosoma X/efectos de los fármacos , Inactivación del Cromosoma X/genética , Administración Oftálmica , Animales , Compuestos de Bencidrilo/administración & dosificación , Compuestos de Bencidrilo/metabolismo , Encéfalo/crecimiento & desarrollo , Estrógenos no Esteroides/administración & dosificación , Estrógenos no Esteroides/metabolismo , Femenino , Intercambio Materno-Fetal , Discapacidad Intelectual Ligada al Cromosoma X/genética , Ratones , Ratones Endogámicos ICR , Fenoles/administración & dosificación , Fenoles/metabolismo , Embarazo , Diferenciación Sexual/genética , Inactivación del Cromosoma X/fisiología
9.
J Toxicol Sci ; 38(2): 245-54, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23535403

RESUMEN

Several studies have shown effects of diesel exhaust (DE) on the central nervous system, but the mechanism is unclear. Fetal mice were exposed to whole DE (contains gases and particles) in an inhalation chamber, and cerebrum gene expression changes were examined by gene assay (microarray and quantitative real-time PCR). By microarray, upregulation of Xist, B-raf and Drwms2 were detected. Especially, mRNA expression of Xist was increased in a concentration-dependent manner in male and female mice. Xist (X-inactive specific transcript) is a major effector of the X-inactivation process, and X-linked genes are highly expressed in brain tissue and consistent with a role in brain developments. By quantitative real-time PCR, Tsix (crucial noncoding antisense partner of Xist) and other X-linked genes (Mecp2, Hprt1, and Sts) were examined; Tsix was upregulated, and other X-linked genes were unaffected in the male and female mice. Our findings suggest that exposure to DE increases Xist and Tsix gene expression in utero without influencing X-linked gene expression. An examination of Xist gene expression changes may provide an important biomarker for DE-induced effects. The possibility of avoiding X-chromosome inactivation (XCI) mechanisms by minimizing exposure to DE is expected.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/embriología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Exposición por Inhalación/efectos adversos , Exposición Materna/efectos adversos , Intercambio Materno-Fetal/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/etiología , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo , Emisiones de Vehículos/toxicidad , Inactivación del Cromosoma X/efectos de los fármacos , Inactivación del Cromosoma X/genética , Animales , Relación Dosis-Respuesta a Droga , Femenino , Perfilación de la Expresión Génica/métodos , Masculino , Ratones , Ratones Endogámicos ICR , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Matrices Tisulares
10.
Reproduction ; 145(1): 9-17, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23104973

RESUMEN

During initial development, both X chromosomes are active in females, and one of them must be silenced at the appropriate time in order to dosage compensate their gene expression levels to male counterparts. Silencing involves epigenetic mechanisms, including histone deacetylation. Major X chromosome inactivation (XCI) in bovine occurs between hatching and implantation, although in vitro culture conditions might disrupt the silencing process, increasing or decreasing X-linked gene expression. In this study, we aimed to address the roles of histone deacetylase inhibition by trichostatin A (TSA) on female preimplantation development. We tested the hypothesis that by enhancing histone acetylation, TSA would increase the percentage of embryos achieving 16-cell stage, reducing percentage of embryos blocked at 8-cell stage, and interfere with XCI in IVF embryos. We noticed that after TSA treatment, acetylation levels in individual blastomeres of 8-16 cell embryos were increased twofold on treated embryos, and the same was detected for blastocysts. Changes among blastomere levels within the same embryo were diminished on TSA group, as low-acetylated blastomeres were no longer detected. The percentage of embryos that reached the 5th cleavage cycle 118 h after IVF, analyzed by Hoechst staining, remained unaltered after TSA treatment. Then, we assessed XIST and G6PD expression in individual female bovine blastocysts by quantitative real-time PCR. Even though G6PD expression remained unaltered after TSA exposure, XIST expression was eightfold decreased, and we also detected a major decrease in the percentage of blastocysts expressing detectable XIST levels after TSA treatment. Based on these results, we conclude that HDAC is involved on XCI process in bovine embryos, and its inhibition might delay X chromosome silencing and attenuate aberrant XIST expression described for IVF embryos.


Asunto(s)
Blastocisto/metabolismo , Bovinos/embriología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , ARN Largo no Codificante/metabolismo , Animales , Blastocisto/efectos de los fármacos , Bovinos/metabolismo , Células Cultivadas , Femenino , Fertilización In Vitro/métodos , Glucosafosfato Deshidrogenasa/metabolismo , Histona Desacetilasas/metabolismo , Técnicas In Vitro , Modelos Animales , Inactivación del Cromosoma X/efectos de los fármacos
11.
Mol Ther ; 20(10): 1953-67, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22760542

RESUMEN

Induced pluripotent stem cells (iPSCs) with potential for therapeutic applications can be derived from somatic cells via ectopic expression of a set of limited and defined transcription factors. However, due to risks of random integration of the reprogramming transgenes into the host genome, the low efficiency of the process, and the potential risk of virally induced tumorigenicity, alternative methods have been developed to generate pluripotent cells using nonintegrating systems, albeit with limited success. Here, we show that c-KIT+ human first-trimester amniotic fluid stem cells (AFSCs) can be fully reprogrammed to pluripotency without ectopic factors, by culture on Matrigel in human embryonic stem cell (hESC) medium supplemented with the histone deacetylase inhibitor (HDACi) valproic acid (VPA). The cells share 82% transcriptome identity with hESCs and are capable of forming embryoid bodies (EBs) in vitro and teratomas in vivo. After long-term expansion, they maintain genetic stability, protein level expression of key pluripotency factors, high cell-division kinetics, telomerase activity, repression of X-inactivation, and capacity to differentiate into lineages of the three germ layers, such as definitive endoderm, hepatocytes, bone, fat, cartilage, neurons, and oligodendrocytes. We conclude that AFSC can be utilized for cell banking of patient-specific pluripotent cells for potential applications in allogeneic cellular replacement therapies, pharmaceutical screening, and disease modeling.


Asunto(s)
Líquido Amniótico/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ácido Valproico/farmacología , Líquido Amniótico/citología , Diferenciación Celular , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Genoma Humano , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipificación , Cinética , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fenotipo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Análisis de Secuencia de ADN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma , Transgenes , Inactivación del Cromosoma X/efectos de los fármacos
12.
Cell Stem Cell ; 11(1): 75-90, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22770242

RESUMEN

Although human induced pluripotent stem cells (hiPSCs) have enormous potential in regenerative medicine, their epigenetic variability suggests that some lines may not be suitable for human therapy. There are currently few benchmarks for assessing quality. Here we show that X-inactivation markers can be used to separate hiPSC lines into distinct epigenetic classes and that the classes are phenotypically distinct. Loss of XIST expression is strongly correlated with upregulation of X-linked oncogenes, accelerated growth rate in vitro, and poorer differentiation in vivo. Whereas differences in X-inactivation potential result in epigenetic variability of female hiPSC lines, male hiPSC lines generally resemble each other and do not overexpress the oncogenes. Neither physiological oxygen levels nor HDAC inhibitors offer advantages to culturing female hiPSC lines. We conclude that female hiPSCs may be epigenetically less stable in culture and caution that loss of XIST may result in qualitatively less desirable stem cell lines.


Asunto(s)
Perfilación de la Expresión Génica , Genes Relacionados con las Neoplasias/genética , Células Madre Pluripotentes Inducidas/metabolismo , Neoplasias/genética , Caracteres Sexuales , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Cromosomas Humanos X/genética , Femenino , Genoma Humano/genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Masculino , Ratones , Neoplasias/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxígeno/farmacología , ARN Largo no Codificante , ARN no Traducido/genética , ARN no Traducido/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Inactivación del Cromosoma X/efectos de los fármacos , Inactivación del Cromosoma X/genética
13.
Alcohol Clin Exp Res ; 36(8): 1325-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22375556

RESUMEN

BACKGROUND: All female mammals with 2 X chromosomes balance gene expression with males having only 1 X by inactivating one of their X chromosomes (X chromosome inactivation [XCI]). Analysis of XCI in females offers the opportunity to investigate both X-linked genetic factors and early embryonic development that may contribute to alcoholism. Increases in the prevalence of skewing of XCI in women with alcoholism could implicate biological risk factors. METHODS: The pattern of XCI was examined in DNA isolated in blood from 44 adult women meeting DSM-IV criteria for an alcohol use disorder and 45 control women with no known history of alcohol abuse or dependence. XCI status was determined by analyzing digested and undigested polymerase chain reaction (PCR) products of the polymorphic androgen receptor (AR) gene located on the X chromosome. Subjects were categorized into 3 groups based upon the degree of XCI skewness: random (50:50 to 64:36%), moderately skewed (65:35 to 80:20%), and highly skewed (>80:20%). RESULTS: XCI status from informative women with alcoholism was found to be random in 59% (n = 26), moderately skewed in 27% (n = 12), or highly skewed in 14% (n = 6). Control subjects showed 60, 29, and 11%, respectively. The distribution of skewed XCI observed among women with alcoholism did not differ statistically from that of control subjects (χ(2) test = 0.14, 2 df, p = 0.93). CONCLUSIONS: Our data did not support an increase in XCI skewness among women with alcoholism or implicate early developmental events associated with embryonic cell loss or unequal (nonrandom) expression of X-linked gene(s) or defects in alcoholism among women.


Asunto(s)
Alcoholismo/genética , Inactivación del Cromosoma X/efectos de los fármacos , Adulto , ADN/biosíntesis , ADN/genética , Manual Diagnóstico y Estadístico de los Trastornos Mentales , Desarrollo Embrionario/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Genes Ligados a X/efectos de los fármacos , Humanos , Masculino , Reacción en Cadena de la Polimerasa , Embarazo , Receptores Androgénicos/genética , Factores de Riesgo
14.
Proc Natl Acad Sci U S A ; 106(4): 1122-7, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19164542

RESUMEN

Dicer is central to the RNA interference (RNAi) pathway, because it is required for processing of double-stranded RNA (dsRNA) precursors into small RNA effector molecules. In principle, any long dsRNA could serve as a substrate for Dicer. The X inactive specific transcript (Xist) is an untranslated RNA that is required for dosage compensation in mammals. It coats and silences 1 of the 2 X chromosomes in female cells and initiates a chromosomewide change in chromatin structure that includes the recruitment of Polycomb proteins, but it is largely unknown how Xist RNA mediates these processes. To investigate a potential link between the RNAi pathway and X inactivation, we generated and analyzed Dicer-deficient embryonic stem (ES) cells. In the absence of Dicer, coating by Xist RNA, initiation of silencing, and recruitment of Polycomb proteins occur normally. Dicer ablation had modest effects on the steady-state levels of spliced Xist RNA. Together our data indicate that the RNAi machinery is not essential for the initiation of X inactivation.


Asunto(s)
ARN Helicasas DEAD-box/deficiencia , Endorribonucleasas/deficiencia , Inactivación del Cromosoma X/genética , Animales , Cruzamientos Genéticos , ARN Helicasas DEAD-box/metabolismo , Doxiciclina/farmacología , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Endorribonucleasas/metabolismo , Femenino , Silenciador del Gen/efectos de los fármacos , Genes Ligados a X , Endogamia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , Proteínas del Grupo Polycomb , ARN Largo no Codificante , ARN no Traducido/metabolismo , Proteínas Represoras/metabolismo , Ribonucleasa III , Tetraciclina/farmacología , Cromosoma X/genética , Inactivación del Cromosoma X/efectos de los fármacos
15.
Reproduction ; 135(6): 815-28, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18304987

RESUMEN

The poor outcome of somatic cell nuclear transfer (SCNT) is thought to be a consequence of incomplete reprogramming of the donor cell. The objective of this study was to investigate the effects of treatment with S-adenosylhomocysteine (SAH) a DNA demethylation agent, on DNA methylation levels and X-chromosome inactivation status of bovine female fibroblast donor cells and the subsequent impact on developmental potential after SCNT. Compared with non-treated controls, the cells treated with SAH revealed (i) significantly (P<0.05) reduced global DNA methylation, (ii) significantly (approximately 1.5-fold) increased telomerase activity, (iii) diminished distribution signals of methylated histones H3-3mK9 and H3-3mK27 on the presumptive inactive X-chromosome (Xi), (iv) alteration in the replication pattern of the Xi, and (v) elevation of transcript levels for X-chromosome linked genes, ANT3, MECP2, XIAP, XIST, and HPRT. SCNT embryos produced with SAH-treated donor cells compared with those derived from untreated donor cells revealed (i) similar cleavage frequencies, (ii) significant elevation in the frequencies of development of cleaved embryos to hatched blastocyst stage, and (iii) 1.5-fold increase in telomerase activity. We concluded that SAH induces global DNA demethylation that partially reactivates the Xi, and that a hypomethylated genome may facilitate the nuclear reprogramming process.


Asunto(s)
Fibroblastos/metabolismo , Técnicas de Transferencia Nuclear , S-Adenosilhomocisteína/farmacología , Inactivación del Cromosoma X/efectos de los fármacos , 5-Metilcitosina/análisis , Animales , Bovinos , Reprogramación Celular , Metilación de ADN , Desarrollo Embrionario/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/ultraestructura , Histonas/análisis , Histonas/metabolismo , Metafase , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Telomerasa/metabolismo , Transcripción Genética
17.
Biochem Biophys Res Commun ; 337(3): 875-80, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16213462

RESUMEN

ATM and ATR are well documented for their roles in maintaining the integrity of genomic DNA by responding to DNA damage and preparing the cell for repair. Since ATM and ATR have been reported to exist in complexes with histone deacetylases, we asked whether Atm and Atr might also uphold gene silencing by heterochromatin. We show that the Atm/Atr inhibitor 2-aminopurine causes the inactive X chromosome to accumulate abnormal chromatin and undergo unwanted gene reactivation. We provide evidence that this gene expression from the inactive X chromosome is not a byproduct of the accumulation of DNA breaks. Individually inhibiting Atm and Atr by either small interfering RNA or the expression of dominant-negative ATM and ATR constructs also compromised X-inactivation. Atm and Atr, therefore, not only function in responding to DNA damage but perhaps also are involved in gene silencing via the maintenance of heterochromatin.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Daño del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Fibroblastos/fisiología , Silenciador del Gen/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Inactivación del Cromosoma X/fisiología , Cromosoma X/genética , 2-Aminopurina/farmacología , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular , Daño del ADN/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Rayos gamma , Regulación de la Expresión Génica/genética , Silenciador del Gen/efectos de los fármacos , Silenciador del Gen/efectos de la radiación , Heterocromatina/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Cromosoma X/efectos de los fármacos , Cromosoma X/efectos de la radiación , Inactivación del Cromosoma X/efectos de los fármacos , Inactivación del Cromosoma X/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA