Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Hum Mol Genet ; 29(19): 3285-3295, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-32977341

RESUMEN

Prader-Willi syndrome (PWS) is characterized by neonatal hypotonia, developmental delay and hyperphagia/obesity. This disorder is caused by the absence of paternally expressed gene products from chromosome 15q11-q13. We previously demonstrated that knocking out ZNF274, a Kruppel-associated box-A-domain zinc finger protein capable of recruiting epigenetic machinery to deposit the H3K9me3 repressive histone modification, can activate expression from the normally silent maternal allele of SNORD116 in neurons derived from PWS induced pluripotent stem cells (iPSCs). However, ZNF274 has many other targets in the genome in addition to SNORD116. Depleting ZNF274 will surely affect the expression of other important genes and disrupt other pathways. Here, we used CRISPR/Cas9 to delete ZNF274 binding sites at the SNORD116 locus to determine whether activation of the maternal copy of SNORD116 could be achieved without altering ZNF274 protein levels. We obtained similar activation of gene expression from the normally silenced maternal allele in neurons derived from PWS iPSCs, compared with ZNF274 knockout, demonstrating that ZNF274 is directly involved in the repression of SNORD116. These results suggest that interfering with ZNF274 binding at the maternal SNORD116 locus is a potential therapeutic strategy for PWS.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Factores de Transcripción de Tipo Kruppel/metabolismo , Neuronas/patología , Síndrome de Prader-Willi/patología , ARN Mensajero Almacenado/genética , ARN Nucleolar Pequeño/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Neuronas/metabolismo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo
2.
Hum Mol Genet ; 27(3): 505-515, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29228278

RESUMEN

Prader-Willi syndrome (PWS) is characterized by neonatal hypotonia, developmental delay and hyperphagia/obesity and is caused by the absence of paternal contribution to chromosome 15q11-q13. Using induced pluripotent stem cell (iPSC) models of PWS, we previously discovered an epigenetic complex that is comprised of the zinc-finger protein ZNF274 and the SET domain bifurcated 1 (SETDB1) histone H3 lysine 9 (H3K9) methyltransferase and that silences the maternal alleles at the PWS locus. Here, we have knocked out ZNF274 and rescued the expression of silent maternal alleles in neurons derived from PWS iPSC lines, without affecting DNA methylation at the PWS-Imprinting Center (PWS-IC). This suggests that the ZNF274 complex is a separate imprinting mark that represses maternal PWS gene expression in neurons and is a potential target for future therapeutic applications to rescue the PWS phenotype.


Asunto(s)
Impresión Genómica/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Síndrome de Prader-Willi/metabolismo , Alelos , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Metilación de ADN/genética , Epigénesis Genética/genética , Impresión Genómica/fisiología , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Noqueados , Síndrome de Prader-Willi/genética
3.
Hum Mol Genet ; 23(17): 4674-85, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24760766

RESUMEN

Prader-Willi syndrome (PWS), a disorder of genomic imprinting, is characterized by neonatal hypotonia, hypogonadism, small hands and feet, hyperphagia and obesity in adulthood. PWS results from the loss of paternal copies of the cluster of SNORD116 C/D box snoRNAs and their host transcript, 116HG, on human chromosome 15q11-q13. We have investigated the mechanism of repression of the maternal SNORD116 cluster and 116HG. Here, we report that the zinc-finger protein ZNF274, in association with the histone H3 lysine 9 (H3K9) methyltransferase SETDB1, is part of a complex that binds to the silent maternal but not the active paternal alleles. Knockdown of SETDB1 in PWS-specific induced pluripotent cells (iPSCs) causes a decrease in the accumulation of H3K9 trimethylation (H3K9me3) at 116HG and corresponding accumulation of the active chromatin mark histone H3 lysine 4 dimethylation (H3K4me2). We also show that upon knockdown of SETDB1 in PWS-specific iPSCs, expression of maternally silenced 116HG RNA is partially restored. SETDB1 knockdown in PWS iPSCs also disrupts DNA methylation at the PWS-IC where a decrease in 5-methylcytosine is observed in association with a concomitant increase in 5-hydroxymethylcytosine. This observation suggests that the ZNF274/SETDB1 complex bound to the SNORD116 cluster may protect the PWS-IC from DNA demethylation during early development. Our findings reveal novel epigenetic mechanisms that function to repress the maternal 15q11-q13 region.


Asunto(s)
Técnicas de Silenciamiento del Gen , Células Madre Pluripotentes Inducidas/metabolismo , Familia de Multigenes , Síndrome de Prader-Willi/genética , Proteína Metiltransferasas/genética , ARN Nucleolar Pequeño/genética , Activación Transcripcional , Alelos , Línea Celular , Metilación de ADN/genética , Epigénesis Genética , Femenino , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Síndrome de Prader-Willi/patología
4.
Hum Mol Genet ; 23(9): 2364-73, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24363065

RESUMEN

Prader-Willi syndrome (PWS) and Angelman syndrome (AS) are two neurodevelopmental disorders most often caused by deletions of the same region of paternally inherited and maternally inherited human chromosome 15q, respectively. AS is a single gene disorder, caused by the loss of function of the ubiquitin ligase E3A (UBE3A) gene, while PWS is still considered a contiguous gene disorder. Rare individuals with PWS who carry atypical microdeletions on chromosome 15q have narrowed the critical region for this disorder to a 108 kb region that includes the SNORD116 snoRNA cluster and the Imprinted in Prader-Willi (IPW) non-coding RNA. Here we report the derivation of induced pluripotent stem cells (iPSCs) from a PWS patient with an atypical microdeletion that spans the PWS critical region. We show that these iPSCs express brain-specific portions of the transcripts driven by the PWS imprinting center, including the UBE3A antisense transcript (UBE3A-ATS). Furthermore, UBE3A expression is imprinted in most of these iPSCs. These data suggest that UBE3A imprinting in neurons only requires UBE3A-ATS expression, and no other neuron-specific factors. These data also suggest that a boundary element lying within the PWS critical region prevents UBE3A-ATS expression in non-neural tissues.


Asunto(s)
Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Eliminación de Secuencia/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Síndrome de Angelman/genética , Síndrome de Angelman/metabolismo , Línea Celular , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Proc Natl Acad Sci U S A ; 107(41): 17668-73, 2010 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-20876107

RESUMEN

Angelman syndrome (AS) and Prader-Willi syndrome (PWS) are neurodevelopmental disorders of genomic imprinting. AS results from loss of function of the ubiquitin protein ligase E3A (UBE3A) gene, whereas the genetic defect in PWS is unknown. Although induced pluripotent stem cells (iPSCs) provide invaluable models of human disease, nuclear reprogramming could limit the usefulness of iPSCs from patients who have AS and PWS should the genomic imprint marks be disturbed by the epigenetic reprogramming process. Our iPSCs derived from patients with AS and PWS show no evidence of DNA methylation imprint erasure at the cis-acting PSW imprinting center. Importantly, we find that, as in normal brain, imprinting of UBE3A is established during neuronal differentiation of AS iPSCs, with the paternal UBE3A allele repressed concomitant with up-regulation of the UBE3A antisense transcript. These iPSC models of genomic imprinting disorders will facilitate investigation of the AS and PWS disease processes and allow study of the developmental timing and mechanism of UBE3A repression in human neurons.


Asunto(s)
Síndrome de Angelman/genética , Diferenciación Celular/fisiología , Impresión Genómica/genética , Modelos Biológicos , Células Madre Pluripotentes/fisiología , Síndrome de Prader-Willi/genética , Cartilla de ADN/genética , Electrofisiología , Humanos , Inmunohistoquímica , Neuronas/fisiología , Reacción en Cadena de la Polimerasa , Ubiquitina-Proteína Ligasas/genética
6.
Stem Cell Res ; 69: 103086, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37004447

RESUMEN

Our goal is to demonstrate and characterize acute glucocorticoid transcriptome response in human embryonic stem cell (hESC) derived neural cultures. Toward this, we confirmed the differentiation of hESC lines H9 and H1 into post-mitotic neurons and astrocytes, in addition to the expressions of glucocorticoid receptor (GR) protein, and the GR co-chaperone FK506 binding protein 51 (FKBP5). In a series of experiments in hESC-derived neural cultures treated with dexamethasone (Dex) for 6 h, glucocorticoid hormone (GH) response was detected through the transcriptional upregulation of GH-responsive genes, FKBP5 and PER1. Both genes responded to Dex treatment in a dose-dependent fashion, and FKBP5 protein was significantly upregulated after a 12-hour Dex exposure. We additionally examined the transcriptome-wide effects of acute GH exposure in hESC-derived cultures and identified FKBP5 as the most highly up-regulated gene. We identified 30 additional differentially expressed (DE) genes common to cultures derived from both H9 and H1 hESCs whose expression levels changed in both lines with similar magnitudes and direction.


Asunto(s)
Glucocorticoides , Células Madre Embrionarias Humanas , Humanos , Glucocorticoides/farmacología , Células Madre Embrionarias Humanas/metabolismo , Dexametasona/farmacología , Transcriptoma , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
7.
J Cell Biochem ; 112(2): 365-73, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21268055

RESUMEN

Epigenetic mechanisms play essential roles in mammalian neurodevelopment and genetic mutations or chromosomal deletions or duplications of epigenetically regulated loci or pathways result in several important human neurodevelopmental disorders. Postnatal mammalian neurons have among the most structured and dynamic nuclear organization of any cell type. Human chromosome 15q11-13 is an imprinted locus required for normal neurodevelopment and is regulated by a plethora of epigenetic mechanisms in neurons, including multiple noncoding RNAs, parentally imprinted transcription and histone modifications, large-scale chromatin decondensation, and homologous pairing in mature neurons of the mammalian brain. Here, we describe the multiple epigenetic layers regulating 15q11-13 gene expression and chromatin dynamics in neurons and propose a model of how noncoding RNAs may influence the unusual neuronal chromatin structure and dynamics at this locus. We also discuss the need for improved neuronal cell culture systems that model human 15q11-13 and other neurodevelopmental disorders with epigenetic bases in order to test the mechanisms of chromatin dynamics and nuclear organization in neurons. Induced pluripotent stem cells and other stem cell technologies hold promise for improved understanding of and therapeutic interventions for multiple human neurodevelopmental disorders.


Asunto(s)
Cromatina/genética , Epigénesis Genética/genética , Animales , Impresión Genómica/genética , Humanos , Modelos Biológicos , Neuronas/metabolismo , ARN Nucleolar Pequeño/genética , ARN no Traducido/genética
8.
Neurobiol Dis ; 39(1): 13-20, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20304067

RESUMEN

Human chromosome 15q11-q13 is subject to regulation by genomic imprinting, an epigenetic process by which genes are expressed in a parent-of-origin specific manner. Three neurodevelopmental disorders, Prader-Willi syndrome, Angelman syndrome, and 15q duplication syndrome, result from aberrant expression of imprinted genes in this region. Here, we review the current literature pertaining to mouse models and recently identified patients with atypical deletions, which shed light on the epigenetic regulation of the chromosome 15q11-q13 subregion and the genes that are responsible for the phenotypic outcomes of these disorders.


Asunto(s)
Cromosomas Humanos Par 15/genética , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/metabolismo , Predisposición Genética a la Enfermedad/genética , Impresión Genómica/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Animales , Discapacidades del Desarrollo/patología , Humanos , Discapacidad Intelectual/patología
9.
Neurogenetics ; 9(4): 227-35, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18791750

RESUMEN

The recent discovery of genomic reprogramming of human somatic cells into induced pluripotent stem cells offers an innovative and relevant approach to the study of human genetic and neurogenetic diseases. By reprogramming somatic cells from patient samples, cell lines can be isolated that self-renew indefinitely and have the potential to develop into multiple different tissue lineages. Additionally, the rapid progress of research on human embryonic stem cells has led to the development of sophisticated in vitro differentiation protocols that closely mimic mammalian development. In particular, there have been significant advances in differentiating human pluripotent stem cells into defined neuronal types. Here, we summarize the experimental approaches employed in the rapidly evolving area of somatic cell reprogramming and the methodologies for differentiating human pluripotent cells into neurons. We also discuss how the availability of patient-specific fibroblasts offers a unique opportunity for studying and modeling the effects of specific gene defects on human neuronal development in vitro and for testing small molecules or other potential therapies for the relevant neurogenetic disorders.


Asunto(s)
Modelos Neurológicos , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/patología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/patología , Diferenciación Celular/genética , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Vectores Genéticos , Humanos , Técnicas In Vitro , Modelos Genéticos , Neuronas/metabolismo , Neuronas/patología
10.
Nucleic Acids Res ; 33(13): 3976-84, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16027444

RESUMEN

Angelman syndrome (AS) is a neurogenetic disorder characterized by severe mental retardation, 'puppet-like' ataxic gait with jerky arm movements, seizures, EEG abnormalities, hyperactivity and bouts of inappropriate laughter. Individuals with AS fail to inherit a normal active maternal copy of the gene encoding ubiquitin protein ligase E3A (UBE3A). UBE3A is transcribed predominantly from the maternal allele in brain, but is expressed from both alleles in most other tissues. It has been proposed that brain-specific silencing of the paternal UBE3A allele is mediated by a large (>500 kb) paternal non-coding antisense transcript (UBE3A-ATS). There are several other examples of imprinting regulation involving antisense transcripts that share two main properties: (i) the sense transcript is repressed by antisense and (ii) the interaction between sense and antisense occurs in cis. We show here that, in a mouse model of AS, maternal transmission of Ube3a mutation leads to increased expression of the paternal Ube3a-ATS, suggesting that the antisense is modulated by sense rather than the reciprocal mode of regulation. Our observation that Ube3a regulates expression of Ube3a-ATS in trans is in contrast to the other cases of sense-antisense epigenetic cis-interactions and argues against a major role for Ube3a-ATS in the imprinting of Ube3a.


Asunto(s)
Síndrome de Angelman/genética , Impresión Genómica , ARN sin Sentido/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Encéfalo/metabolismo , Femenino , Ratones , Mutación , ARN sin Sentido/biosíntesis , Ubiquitina-Proteína Ligasas/biosíntesis , Regulación hacia Arriba
12.
Gene ; 383: 93-8, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16996702

RESUMEN

UBE3A, the gene associated with Angelman syndrome, is part of a cluster of genes in the human chromosome 15q11-q13/mouse chromosome 7C region, that is subject to genomic imprinting. In human and mouse brain, UBE3A is expressed predominantly from the maternal allele, and the paternal allele is silenced. A current model concerning the evolution of genomic imprinting, the parental conflict hypothesis, posits that this epigenetic phenomenon is restricted to eutherian mammals. It has been recently reported, however, that several chicken orthologues of mammalian imprinted loci display DNA replication asynchrony, a property of imprinted genes. A separate group also reported monoallelic expression of chicken IGF2 in developing chicken embryos. These observations could suggest that genomic imprinting may occur in chicken. We have assembled the predicted mRNA consensus sequence for the chicken UBE3A gene using published ESTs. We report a high degree of homology with the human UBE3A at the nucleotide and protein levels, as well as a highly conserved genomic organization. Biallelic expression of UBE3A is observed in embryonic chicken brain and limb, indicating that UBE3A is not subject to genomic imprinting in chicken.


Asunto(s)
Pollos/genética , Ubiquitina-Proteína Ligasas/genética , Alelos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Embrión de Pollo , Secuencia de Consenso , ADN/genética , Etiquetas de Secuencia Expresada , Femenino , Expresión Génica , Genoma , Impresión Genómica , Humanos , Masculino , Modelos Genéticos , Datos de Secuencia Molecular , ARN Mensajero/genética , Homología de Secuencia de Aminoácido , Especificidad de la Especie
13.
Nucleic Acids Res ; 32(11): 3480-92, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15226413

RESUMEN

Most cases of Angelman syndrome (AS) result from loss or inactivation of ubiquitin protein ligase 3A (UBE3A), a gene displaying maternal-specific expression in brain. Epigenetic silencing of the paternal UBE3A allele in brain appears to be mediated by a non-coding UBE3A antisense (UBE3A-ATS). In human, UBE3A-ATS extends approximately 450 kb to UBE3A from the small nuclear ribonucleoprotein N (SNURF/SNRPN) promoter region that contains a cis-acting imprinting center (IC). The concept of a single large antisense transcript is difficult to reconcile with the observation that SNURF/SNRPN shows a ubiquitous pattern of expression while the more distal part of UBE3A-ATS, which overlaps UBE3A, is brain specific. To address this problem, we examined murine transcripts initiating from several alternative exons dispersed within a 500 kb region upstream of Snurf/Snrpn. Similar to Ube3a-ATS, these upstream (U) exon-containing transcripts are expressed at neuronal stages of differentiation in a cell culture model of neurogenesis. These findings suggest the novel hypothesis that brain-specific transcription of Ube3a-ATS is regulated by the U exons rather than Snurf/Snrpn exon 1 as previously suggested from human studies. In support of this hypothesis, we describe U-Ube3a-ATS transcripts where U exons are spliced to Ube3a-ATS with the exclusion of Snurf-Snrpn. We also show that the murine U exons have arisen by genomic duplication of segments that include elements of the IC, suggesting that the brain specific silencing of Ube3a is due to multiple alternatively spliced IC-Ube3a-ATS transcripts.


Asunto(s)
Empalme Alternativo , Encéfalo/metabolismo , Exones , Impresión Genómica , Ratones/genética , ARN sin Sentido/genética , Ubiquitina-Proteína Ligasas/genética , Alelos , Animales , Autoantígenos , Secuencia de Bases , Diferenciación Celular , Línea Celular Tumoral , Silenciador del Gen , Intrones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/genética , ARN sin Sentido/metabolismo , ARN Nucleolar Pequeño/metabolismo , Ratas , Ribonucleoproteínas Nucleares Pequeñas/genética , Alineación de Secuencia , Proteínas Nucleares snRNP
14.
Artículo en Inglés | MEDLINE | ID: mdl-27168766

RESUMEN

BACKGROUND: Eukaryotic genome duplication starts at discrete sequences (replication origins) that coordinate cell cycle progression, ensure genomic stability and modulate gene expression. Origins share some sequence features, but their activity also responds to changes in transcription and cellular differentiation status. RESULTS: To identify chromatin states and histone modifications that locally mark replication origins, we profiled origin distributions in eight human cell lines representing embryonic and differentiated cell types. Consistent with a role of chromatin structure in determining origin activity, we found that cancer and non-cancer cells of similar lineages exhibited highly similar replication origin distributions. Surprisingly, our study revealed that DNase hypersensitivity, which often correlates with early replication at large-scale chromatin domains, did not emerge as a strong local determinant of origin activity. Instead, we found that two distinct sets of chromatin modifications exhibited strong local associations with two discrete groups of replication origins. The first origin group consisted of about 40,000 regions that actively initiated replication in all cell types and preferentially colocalized with unmethylated CpGs and with the euchromatin markers, H3K4me3 and H3K9Ac. The second group included origins that were consistently active in cells of a single type or lineage and preferentially colocalized with the heterochromatin marker, H3K9me3. Shared origins replicated throughout the S-phase of the cell cycle, whereas cell-type-specific origins preferentially replicated during late S-phase. CONCLUSIONS: These observations are in line with the hypothesis that differentiation-associated changes in chromatin and gene expression affect the activation of specific replication origins.

16.
Cell Stem Cell ; 16(5): 533-46, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25921272

RESUMEN

Human pluripotent stem cells (hPSCs) display extensive epigenetic instability, particularly on the X chromosome. In this study, we show that, in hPSCs, the inactive X chromosome has a specific heterochromatin landscape that predisposes it to erosion of X chromosome inactivation (XCI), a process that occurs spontaneously in hPSCs. Heterochromatin remodeling and gene reactivation occur in a non-random fashion and are confined to specific H3K27me3-enriched domains, leaving H3K9me3-marked regions unaffected. Using single-cell monitoring of XCI erosion, we show that this instability only occurs in pluripotent cells. We also provide evidence that loss of XIST expression is not the primary cause of XCI instability and that gene reactivation from the inactive X (Xi) precedes loss of XIST coating. Notably, expression and coating by the long non-coding RNA XACT are early events in XCI erosion and, therefore, may play a role in mediating this process.


Asunto(s)
Cromosomas Humanos X/genética , Histonas/metabolismo , Células Madre Pluripotentes/fisiología , ARN Largo no Codificante/metabolismo , Línea Celular , Ensamble y Desensamble de Cromatina , Represión Epigenética , Heterocromatina/metabolismo , Histonas/genética , Humanos , ARN Largo no Codificante/genética , Transcripción Genética , Inactivación del Cromosoma X
17.
Mol Autism ; 5: 44, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25694803

RESUMEN

BACKGROUND: Duplications of the chromosome 15q11-q13.1 region are associated with an estimated 1 to 3% of all autism cases, making this copy number variation (CNV) one of the most frequent chromosome abnormalities associated with autism spectrum disorder (ASD). Several genes located within the 15q11-q13.1 duplication region including ubiquitin protein ligase E3A (UBE3A), the gene disrupted in Angelman syndrome (AS), are involved in neural function and may play important roles in the neurobehavioral phenotypes associated with chromosome 15q11-q13.1 duplication (Dup15q) syndrome. METHODS: We have generated induced pluripotent stem cell (iPSC) lines from five different individuals containing CNVs of 15q11-q13.1. The iPSC lines were differentiated into mature, functional neurons. Gene expression across the 15q11-q13.1 locus was compared among the five iPSC lines and corresponding iPSC-derived neurons using quantitative reverse transcription PCR (qRT-PCR). Genome-wide gene expression was compared between neurons derived from three iPSC lines using mRNA-Seq. RESULTS: Analysis of 15q11-q13.1 gene expression in neurons derived from Dup15q iPSCs reveals that gene copy number does not consistently predict expression levels in cells with interstitial duplications of 15q11-q13.1. mRNA-Seq experiments show that there is substantial overlap in the genes differentially expressed between 15q11-q13.1 deletion and duplication neurons, Finally, we demonstrate that UBE3A transcripts can be pharmacologically rescued to normal levels in iPSC-derived neurons with a 15q11-q13.1 duplication. CONCLUSIONS: Chromatin structure may influence gene expression across the 15q11-q13.1 region in neurons. Genome-wide analyses suggest that common neuronal pathways may be disrupted in both the Angelman and Dup15q syndromes. These data demonstrate that our disease-specific stem cell models provide a new tool to decipher the underlying cellular and genetic disease mechanisms of ASD and may also offer a pathway to novel therapeutic intervention in Dup15q syndrome.

18.
Epigenetics ; 7(1): 71-82, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22207353

RESUMEN

The de novo DNA methyltransferase DNMT3B functions in establishing DNA methylation patterns during development. DNMT3B missense mutations cause immunodeficiency, centromere instability and facial anomalies (ICF) syndrome. The restriction of Dnmt3b expression to neural progenitor cells, as well as the mild cognitive defects observed in ICF patients, suggests that DNMT3B may play an important role in early neurogenesis. We performed RNAi knockdown of DNMT3B in human embryonic stem cells (hESCs) in order to investigate the mechanistic contribution of DNMT3B to DNA methylation and early neuronal differentiation. While DNMT3B was not required for early neuroepithelium specification, DNMT3B deficient neuroepithelium exhibited accelerated maturation with earlier expression, relative to normal hESCs, of mature neuronal markers (such as NEUROD1) and of early neuronal regional specifiers (such as those for the neural crest). Genome-wide analyses of DNA methylation by MethylC-seq identified novel regions of hypomethylation in the DNMT3B knockdowns along the X chromosome as well as pericentromeric regions, rather than changes to promoters of specific dysregulated genes. We observed a loss of H3K27me3 and the polycomb complex protein EZH2 at the promoters of early neural and neural crest specifier genes during differentiation of DNMT3B knockdown but not normal hESCs. Our results indicate that DNMT3B mediates large-scale methylation patterns in hESCs and that DNMT3B deficiency in the cells alters the timing of their neuronal differentiation and maturation.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Estudio de Asociación del Genoma Completo , Cresta Neural/metabolismo , Neurogénesis , Línea Celular , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Células Madre Embrionarias/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Cresta Neural/citología , Células Madre Pluripotentes/metabolismo , ADN Metiltransferasa 3B
20.
Plasmid ; 55(3): 216-26, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16439020

RESUMEN

RNA interference is a widely used tool for analysis of gene function in mammalian cells. Stable knockdown of specific target genes can be maintained in cell lines and live organisms using vector-based delivery of short hairpins (shRNAs) driven by RNA polymerase III promoters. Here we describe a vector incorporating the human 7SK promoter for shRNA-mediated gene silencing in the P19 embryonic carcinoma stem cell line. Our preliminary experiments with the 7SK shRNA expression vector indicated that its activity could be hindered by random genomic integration. In order to counter this inhibitory mechanism, we inserted a matrix-attached region sequence to generate an episomal vector system. We compared the effects of insertion versus exclusion of the MAR sequence on the shRNA-mediated gene-specific silencing of the beta-tubulin III and Cyclophilin A genes. While the MAR sequence is not strongly correlated with the episomal status of the expression vector, our studies indicate that inclusion of the MAR element significantly enhances the stability of shRNA-mediated gene silencing in the P19 stem cells.


Asunto(s)
Carcinoma Embrionario , ADN Polimerasa III/genética , Silenciador del Gen , Vectores Genéticos , Regiones de Fijación a la Matriz/genética , ARN Interferente Pequeño/genética , Secuencia de Bases , Carcinoma Embrionario/metabolismo , Carcinoma Embrionario/patología , Ciclofilina A/antagonistas & inhibidores , Ciclofilina A/genética , Ciclofilina A/metabolismo , ADN Polimerasa III/antagonistas & inhibidores , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Tubulina (Proteína)/química , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA