Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(40): e2211073119, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36161909

RESUMEN

Mammalian sex chromosomes encode homologous X/Y gene pairs that were retained on the Y chromosome in males and escape X chromosome inactivation (XCI) in females. Inferred to reflect X/Y pair dosage sensitivity, monosomy X is a leading cause of miscarriage in humans with near full penetrance. This phenotype is shared with many other mammals but not the mouse, which offers sophisticated genetic tools to generate sex chromosomal aneuploidy but also tolerates its developmental impact. To address this critical gap, we generated X-monosomic human induced pluripotent stem cells (hiPSCs) alongside otherwise isogenic euploid controls from male and female mosaic samples. Phased genomic variants in these hiPSC panels enable systematic investigation of X/Y dosage-sensitive features using in vitro models of human development. Here, we demonstrate the utility of these validated hiPSC lines to test how X/Y-linked gene dosage impacts a widely used model for human syncytiotrophoblast development. While these isogenic panels trigger a GATA2/3- and TFAP2A/C-driven trophoblast gene circuit irrespective of karyotype, differential expression implicates monosomy X in altered levels of placental genes and in secretion of placental growth factor (PlGF) and human chorionic gonadotropin (hCG). Remarkably, weighted gene coexpression network modules that significantly reflect these changes are also preserved in first-trimester chorionic villi and term placenta. Our results suggest monosomy X may skew trophoblast cell type composition and function, and that the combined haploinsufficiency of the pseudoautosomal region likely plays a key role in these changes.


Asunto(s)
Dosificación de Gen , Células Madre Pluripotentes Inducidas , Trofoblastos , Síndrome de Turner , Animales , Línea Celular , Gonadotropina Coriónica/metabolismo , Cromosomas Humanos X/genética , Femenino , Humanos , Masculino , Ratones , Factor de Crecimiento Placentario/metabolismo , Embarazo , Trofoblastos/metabolismo , Síndrome de Turner/genética
2.
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
3.
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
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.
bioRxiv ; 2024 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-36945647

RESUMEN

Modeling the developmental etiology of viable human aneuploidy can be challenging in rodents due to syntenic boundaries, or primate-specific biology. In humans, monosomy-X (45,X) causes Turner syndrome (TS), altering craniofacial, skeletal, endocrine, and cardiovascular development, which in contrast remain unaffected in 39,X-mice. To learn how human monosomy-X may impact early embryonic development, we turned to human 45,X and isogenic euploid induced pluripotent stem cells (hiPSCs) from male and female mosaic donors. Because neural crest (NC) derived cell types are hypothesized to underpin craniofacial and cardiovascular changes in TS, we performed a highly-powered differential expression study on hiPSC-derived anterior neural crest cells (NCCs). Across three independent isogenic panels, 45,X NCCs show impaired acquisition of PAX7+SOX10+ markers, and disrupted expression of other NCC-specific genes, relative to their isogenic euploid controls. In particular, 45,X NCCs increase cholesterol biosynthesis genes while reducing transcripts that feature 5' terminal oligopyrimidine (TOP) motifs, including those of ribosomal protein and nuclear-encoded mitochondrial genes. Such metabolic pathways are also over-represented in weighted co-expression gene modules that are preserved in monogenic neurocristopathy. Importantly, these gene modules are also significantly enriched in 28% of all TS-associated terms of the human phenotype ontology. Our analysis identifies specific sex-linked genes that are expressed from two copies in euploid males and females alike and qualify as candidate haploinsufficient drivers of TS phenotypes in NC-derived lineages. This study demonstrates that isogenic hiPSC-derived NCC panels representing monosomy-X can serve as a powerful model of early NC development in TS and inform new hypotheses towards its etiology.

6.
Sci Adv ; 10(23): eadj0385, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38848354

RESUMEN

Excess gene dosage from chromosome 21 (chr21) causes Down syndrome (DS), spanning developmental and acute phenotypes in terminal cell types. Which phenotypes remain amenable to intervention after development is unknown. To address this question in a model of DS neurogenesis, we derived trisomy 21 (T21) human induced pluripotent stem cells (iPSCs) alongside, otherwise, isogenic euploid controls from mosaic DS fibroblasts and equipped one chr21 copy with an inducible XIST transgene. Monoallelic chr21 silencing by XIST is near-complete and irreversible in iPSCs. Differential expression reveals that T21 neural lineages and iPSCs share suppressed translation and mitochondrial pathways and activate cellular stress responses. When XIST is induced before the neural progenitor stage, T21 dosage correction suppresses a pronounced skew toward astrogenesis in neural differentiation. Because our transgene remains inducible in postmitotic T21 neurons and astrocytes, we demonstrate that XIST efficiently represses genes even after terminal differentiation, which will empower exploration of cell type-specific T21 phenotypes that remain responsive to chr21 dosage.


Asunto(s)
Diferenciación Celular , Síndrome de Down , Dosificación de Gen , Células Madre Pluripotentes Inducidas , Neurogénesis , ARN Largo no Codificante , Síndrome de Down/genética , Humanos , Neurogénesis/genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , ARN Largo no Codificante/genética , Diferenciación Celular/genética , Cromosomas Humanos Par 21/genética , Neuronas/metabolismo
7.
Curr Biol ; 15(22): 2063-8, 2005 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-16303568

RESUMEN

Hyperactivation of p53 leads to a reduction in tumor formation and an unexpected shortening of life span in two different model systems . The decreased life span occurs with signs of accelerated aging, such as osteoporosis, reduction in body weight, atrophy of organs, decreased stress resistance, and depletion of hematopoietic stem cells. These observations suggest a role for p53 in the determination of life span and the speculation that decreasing p53 activity may result in positive effects on some aging phenotypes . In this report, we show that expression of dominant-negative versions of Drosophila melanogaster p53 in adult neurons extends life span and increases genotoxic stress resistance in the fly. Consistent with this, a naturally occurring allele with decreased p53 activity has been associated with extended survival in humans . Expression of the dominant-negative Drosophila melanogaster p53 constructs does not further increase the extended life span of flies that are calorie restricted, suggesting that a decrease in p53 activity may mediate a component of the calorie-restriction life span-extending pathway in flies.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Longevidad/fisiología , Neuronas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Drosophila melanogaster/metabolismo , Metabolismo Energético/fisiología , Fertilidad/fisiología , Longevidad/genética , Estrés Oxidativo/fisiología
8.
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
9.
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
10.
Methods Mol Biol ; 1353: 45-64, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25520291

RESUMEN

Induced pluripotent stem cell (iPSC) technology has allowed for the invaluable modeling of many genetic disorders including disorders associated with genomic imprinting. Genomic imprinting involves differential DNA and histone methylation and results in allele-specific gene expression. Most of the epigenetic marks in somatic cells are erased and reestablished during the process of reprogramming into iPSCs. Therefore, in generating models of disorders associated with genomic imprinting, it is important to verify that the imprinting status and allele-specific gene expression patterns of the parental somatic cells are maintained in their derivative iPSCs. Here, we describe three techniques: DNA methylation analysis, allele-specific PCR, and RNA FISH, which we use to analyze genomic imprinting in iPSC models of neurogenetic disorders involving copy number variations of the chromosome 15q11-q13 region.


Asunto(s)
Epigénesis Genética , Impresión Genómica , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Genéticos , Síndrome de Prader-Willi/genética , Alelos , Animales , Diferenciación Celular , Células Cultivadas , Variaciones en el Número de Copia de ADN , Metilación de ADN , Cartilla de ADN/síntesis química , Cartilla de ADN/metabolismo , Células Nutrientes/citología , Fibroblastos/citología , Humanos , Hibridación Fluorescente in Situ/métodos , Células Madre Pluripotentes Inducidas/patología , Ratones , Reacción en Cadena de la Polimerasa/métodos , Síndrome de Prader-Willi/diagnóstico , Síndrome de Prader-Willi/patología , ARN/genética , ARN/metabolismo , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
12.
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.

13.
Exp Cell Res ; 298(1): 262-7, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15242780

RESUMEN

The epigenetic states of key regulatory genes must be altered to drive cell fate decisions in differentiating cells. This process must be coupled, at least transiently, to the DNA replication machinery. Only a few genes, however, have been shown to require DNA replication for their activation or repression upon induction of differentiation. We have developed a methodology for examining how gene expression is coupled to cell division during the early stages of differentiation of embryonal carcinoma (EC) cells. Using this approach, we find that the expression of the 5-hydroxytryptamine (serotonin) receptor 2C (Htr2c) is strongly increased in the second division after all-trans retinoic acid addition. We propose that the epigenetic activation of Htr2c in EC cells results from a chromatin remodeling process that requires at least two passages through S phase.


Asunto(s)
Diferenciación Celular/genética , Replicación del ADN/genética , Epigénesis Genética/genética , Regulación del Desarrollo de la Expresión Génica/genética , Células Madre Neoplásicas/metabolismo , Receptor de Serotonina 5-HT2C/genética , Animales , División Celular/genética , Línea Celular , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/genética , Replicación del ADN/efectos de los fármacos , Células Madre de Carcinoma Embrionario , Epigénesis Genética/efectos de los fármacos , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Receptor de Serotonina 5-HT2C/efectos de los fármacos , Fase S/genética , Serotonina/metabolismo , Tretinoina/metabolismo , Tretinoina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA