Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Stem Cell Res ; 69: 103086, 2023 06.
Article in English | MEDLINE | ID: mdl-37004447

ABSTRACT

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.


Subject(s)
Glucocorticoids , Human Embryonic Stem Cells , Humans , Glucocorticoids/pharmacology , Human Embryonic Stem Cells/metabolism , Dexamethasone/pharmacology , Transcriptome , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
2.
Hum Mol Genet ; 29(19): 3285-3295, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32977341

ABSTRACT

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.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Kruppel-Like Transcription Factors/metabolism , Neurons/pathology , Prader-Willi Syndrome/pathology , RNA, Messenger, Stored/genetics , RNA, Small Nucleolar/genetics , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Transcription Factors/genetics , Neurons/metabolism , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/metabolism
3.
Hum Mol Genet ; 27(3): 505-515, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29228278

ABSTRACT

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.


Subject(s)
Genomic Imprinting/genetics , Kruppel-Like Transcription Factors/metabolism , Prader-Willi Syndrome/metabolism , Alleles , Animals , Cells, Cultured , Chromatin Immunoprecipitation , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Genomic Imprinting/physiology , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Prader-Willi Syndrome/genetics
4.
Article in English | MEDLINE | ID: mdl-27168766

ABSTRACT

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.

5.
Cell Stem Cell ; 16(5): 533-46, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25921272

ABSTRACT

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.


Subject(s)
Chromosomes, Human, X/genetics , Histones/metabolism , Pluripotent Stem Cells/physiology , RNA, Long Noncoding/metabolism , Cell Line , Chromatin Assembly and Disassembly , Epigenetic Repression , Heterochromatin/metabolism , Histones/genetics , Humans , RNA, Long Noncoding/genetics , Transcription, Genetic , X Chromosome Inactivation
6.
Hum Mol Genet ; 23(17): 4674-85, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24760766

ABSTRACT

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.


Subject(s)
Gene Knockdown Techniques , Induced Pluripotent Stem Cells/metabolism , Multigene Family , Prader-Willi Syndrome/genetics , Protein Methyltransferases/genetics , RNA, Small Nucleolar/genetics , Transcriptional Activation , Alleles , Cell Line , DNA Methylation/genetics , Epigenesis, Genetic , Female , Gene Silencing , Histone-Lysine N-Methyltransferase , Humans , Kruppel-Like Transcription Factors/genetics , Prader-Willi Syndrome/pathology
7.
Hum Mol Genet ; 23(9): 2364-73, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24363065

ABSTRACT

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.


Subject(s)
Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/metabolism , Sequence Deletion/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Cell Line , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
Mol Autism ; 5: 44, 2014.
Article in English | MEDLINE | ID: mdl-25694803

ABSTRACT

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.

10.
Epigenetics ; 7(1): 71-82, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22207353

ABSTRACT

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.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Genome-Wide Association Study , Neural Crest/metabolism , Neurogenesis , Cell Line , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , Embryonic Stem Cells/metabolism , Female , Gene Knockdown Techniques , Humans , Male , Neural Crest/cytology , Pluripotent Stem Cells/metabolism , DNA Methyltransferase 3B
12.
J Cell Biochem ; 112(2): 365-73, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268055

ABSTRACT

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.


Subject(s)
Chromatin/genetics , Epigenesis, Genetic/genetics , Animals , Genomic Imprinting/genetics , Humans , Models, Biological , Neurons/metabolism , RNA, Small Nucleolar/genetics , RNA, Untranslated/genetics
13.
Proc Natl Acad Sci U S A ; 107(41): 17668-73, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20876107

ABSTRACT

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.


Subject(s)
Angelman Syndrome/genetics , Cell Differentiation/physiology , Genomic Imprinting/genetics , Models, Biological , Pluripotent Stem Cells/physiology , Prader-Willi Syndrome/genetics , DNA Primers/genetics , Electrophysiology , Humans , Immunohistochemistry , Neurons/physiology , Polymerase Chain Reaction , Ubiquitin-Protein Ligases/genetics
15.
Neurobiol Dis ; 39(1): 13-20, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20304067

ABSTRACT

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.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Developmental Disabilities/genetics , Developmental Disabilities/metabolism , Genetic Predisposition to Disease/genetics , Genomic Imprinting/genetics , Intellectual Disability/genetics , Intellectual Disability/metabolism , Animals , Developmental Disabilities/pathology , Humans , Intellectual Disability/pathology
17.
Neurogenetics ; 9(4): 227-35, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18791750

ABSTRACT

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.


Subject(s)
Models, Neurological , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Cell Differentiation/genetics , Cells, Cultured , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Genetic Vectors , Humans , In Vitro Techniques , Models, Genetic , Neurons/metabolism , Neurons/pathology
18.
Gene ; 383: 93-8, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16996702

ABSTRACT

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.


Subject(s)
Chickens/genetics , Ubiquitin-Protein Ligases/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Chick Embryo , Consensus Sequence , DNA/genetics , Expressed Sequence Tags , Female , Gene Expression , Genome , Genomic Imprinting , Humans , Male , Models, Genetic , Molecular Sequence Data , RNA, Messenger/genetics , Sequence Homology, Amino Acid , Species Specificity
19.
Plasmid ; 55(3): 216-26, 2006 May.
Article in English | MEDLINE | ID: mdl-16439020

ABSTRACT

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.


Subject(s)
Carcinoma, Embryonal , DNA Polymerase III/genetics , Gene Silencing , Genetic Vectors , Matrix Attachment Regions/genetics , RNA, Small Interfering/genetics , Base Sequence , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Cyclophilin A/antagonists & inhibitors , Cyclophilin A/genetics , Cyclophilin A/metabolism , DNA Polymerase III/antagonists & inhibitors , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Tubulin/chemistry , Tubulin/genetics , Tubulin/metabolism
20.
Dev Biol ; 286(2): 587-600, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16126194

ABSTRACT

The mouse ortholog of the Prader-Willi/Angelman syndrome imprinted domain contains several paternal-specific transcripts and the maternally expressed gene encoding ubiquitin protein ligase E3A (Ube3a). A Large paternal Non-Coding RNA, encompassing Snurf-Snrpn exons and the Ube3a Antisense Transcript (Ube3a-ATS), has been recently characterized and named here LNCAT. Potential roles of LNCAT in imprinting, gene regulation, and disease are likely but have not been investigated. In order to establish the function(s) of LNCAT, we first determined its in vivo spatio-temporal expression pattern at the cellular level during development and in different adult brain tissues. We show here that LNCAT is developmentally regulated, with transcript variants being specifically expressed through neuronal differentiation in postmitotic neurons. We demonstrate that the LNCAT and Snurf-Snrpn transcripts are independent although they share common exons. We show an absence of expression of LNCAT through gametogenesis and in early embryo excluding a role of LNCAT in the imprint establishment. We also report a range of observations that challenges the widely accepted model of imprinted gene silencing of Ube3a. Although these last data do not completely exclude that the LNCAT variants including "Ube3a-ATS"exons could repress the paternal allele of Ube3a, they do allow us to propose an alternative and consistent model.


Subject(s)
Chromosomes, Mammalian , Gene Expression Regulation, Developmental , Genomic Imprinting , RNA, Untranslated/genetics , Animals , Autoantigens , Embryo, Mammalian , Mice , Mice, Inbred Strains , Nuclear Proteins/genetics , Prader-Willi Syndrome , Ribonucleoproteins, Small Nuclear/genetics , Ubiquitin-Protein Ligases/genetics , snRNP Core Proteins
SELECTION OF CITATIONS
SEARCH DETAIL