Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Chem Neurosci ; 14(22): 3993-4012, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37903506

RESUMO

Copy number variants (CNVs) that delete or duplicate 30 genes within the 16p11.2 genomic region give rise to a range of neurodevelopmental phenotypes with high penetrance in humans. Despite the identification of this small region, the mechanisms by which 16p11.2 CNVs lead to disease are unclear. Relevant models, such as human cortical organoids (hCOs), are needed to understand the human-specific mechanisms of neurodevelopmental disease. We generated hCOs from 17 patients and controls, profiling 167,958 cells with single-cell RNA-sequencing analysis, which revealed neuronal-specific differential expression of genes outside the 16p11.2 region that are related to cell-cell adhesion, neuronal projection growth, and neurodevelopmental disorders. Furthermore, 16p11.2 deletion syndrome organoids exhibited reduced mRNA and protein levels of RBFOX1, a gene that can also harbor CNVs linked to neurodevelopmental phenotypes. We found that the genes previously shown to be regulated by RBFOX1 are also perturbed in organoids from patients with the 16p11.2 deletion syndrome and thus identified a novel link between independent CNVs associated with neuronal development and autism. Overall, this work suggests convergent signaling, which indicates the possibility of a common therapeutic mechanism across multiple rare neuronal diseases.


Assuntos
Deleção Cromossômica , Variações do Número de Cópias de DNA , Humanos , Variações do Número de Cópias de DNA/genética , Encéfalo , Fenótipo , Organoides , Fatores de Processamento de RNA/genética
2.
Proc Natl Acad Sci U S A ; 116(6): 2181-2186, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30674673

RESUMO

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by the loss of function from the maternal allele of UBE3A, a gene encoding an E3 ubiquitin ligase. UBE3A is only expressed from the maternally inherited allele in mature human neurons due to tissue-specific genomic imprinting. Imprinted expression of UBE3A is restricted to neurons by expression of UBE3A antisense transcript (UBE3A-ATS) from the paternally inherited allele, which silences the paternal allele of UBE3A in cis However, the mechanism restricting UBE3A-ATS expression and UBE3A imprinting to neurons is not understood. We used CRISPR/Cas9-mediated genome editing to functionally define a bipartite boundary element critical for neuron-specific expression of UBE3A-ATS in humans. Removal of this element led to up-regulation of UBE3A-ATS without repressing paternal UBE3A However, increasing expression of UBE3A-ATS in the absence of the boundary element resulted in full repression of paternal UBE3A, demonstrating that UBE3A imprinting requires both the loss of function from the boundary element as well as the up-regulation of UBE3A-ATS These results suggest that manipulation of the competition between UBE3A-ATS and UBE3A may provide a potential therapeutic approach for AS.


Assuntos
Cromatina/genética , Impressão Genômica , Neurônios/metabolismo , Ubiquitina-Proteína Ligases/genética , Síndrome de Angelman/genética , Sítios de Ligação , Cromatina/metabolismo , Epistasia Genética , Éxons , Expressão Gênica , Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Ligação Proteica , RNA Antissenso , RNA Longo não Codificante , Deleção de Sequência
3.
Sci Rep ; 6: 25368, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-27146458

RESUMO

Angelman Syndrome (AS) is a rare neurodevelopmental disorder caused by loss of function of the maternally inherited copy of UBE3A, an imprinted gene expressed biallelically in most tissues, but expressed exclusively from the maternal allele in neurons. Active transcription of the neuron-specific long non-coding RNA (lncRNA), UBE3A-ATS, has been shown to silence paternal UBE3A. We hypothesized that alternative splicing factors RBFOX2 and RBFOX1 might mediate splicing changes and result in the transcription of UBE3A-ATS in neurons. We found that RBFOX2 and RBFOX1 both bind to UBE3A-ATS transcript in neurons, but are not required for gene expression and/or neuron-specific processing in the SNURF/SNRPN-UBE3A region. However, we found that depletion of RBFOX2 causes a proliferation phenotype in immature neural cultures, suggesting that RBFOX2 is involved in division versus differentiation decisions in iPSC-derived neural progenitors. Absence of RBFOX2 also altered the expression of some genes that are important for glutamatergic neocortical development and Wnt-Frizzled signalling in mature neuronal cultures. Our data show that while RBFOX1 and RBFOX2 do not mediate neuron-specific processing of UBE3A-ATS, these proteins play important roles in developing neurons and are not completely functionally redundant.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/citologia , Fatores de Processamento de RNA/metabolismo , RNA Longo não Codificante/genética , Proteínas Repressoras/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Fibroblastos/citologia , Impressão Genômica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Neurônios/metabolismo , Fatores de Processamento de RNA/genética , Proteínas Repressoras/genética , Via de Sinalização Wnt
4.
Methods Mol Biol ; 1353: 45-64, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25520291

RESUMO

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.


Assuntos
Epigênese Genética , Impressão Genômica , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Genéticos , Síndrome de Prader-Willi/genética , Alelos , Animais , Diferenciação Celular , Células Cultivadas , Variações do Número de Cópias de DNA , Metilação de DNA , Primers do DNA/síntese química , Primers do DNA/metabolismo , Células Alimentadoras/citologia , Fibroblastos/citologia , Humanos , Hibridização in Situ Fluorescente/métodos , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos , Reação em Cadeia da Polimerase/métodos , Síndrome de Prader-Willi/diagnóstico , Síndrome de Prader-Willi/patologia , RNA/genética , RNA/metabolismo , RNA Nucleolar Pequeno/genética , RNA Nucleolar Pequeno/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
5.
Hum Mol Genet ; 23(9): 2364-73, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24363065

RESUMO

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.


Assuntos
Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Deleção de Sequência/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Síndrome de Angelman/genética , Síndrome de Angelman/metabolismo , Linhagem Celular , Humanos , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Nature ; 501(7465): 58-62, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23995680

RESUMO

Topoisomerases are expressed throughout the developing and adult brain and are mutated in some individuals with autism spectrum disorder (ASD). However, how topoisomerases are mechanistically connected to ASD is unknown. Here we find that topotecan, a topoisomerase 1 (TOP1) inhibitor, dose-dependently reduces the expression of extremely long genes in mouse and human neurons, including nearly all genes that are longer than 200 kilobases. Expression of long genes is also reduced after knockdown of Top1 or Top2b in neurons, highlighting that both enzymes are required for full expression of long genes. By mapping RNA polymerase II density genome-wide in neurons, we found that this length-dependent effect on gene expression was due to impaired transcription elongation. Interestingly, many high-confidence ASD candidate genes are exceptionally long and were reduced in expression after TOP1 inhibition. Our findings suggest that chemicals and genetic mutations that impair topoisomerases could commonly contribute to ASD and other neurodevelopmental disorders.


Assuntos
Transtorno Autístico/genética , DNA Topoisomerases Tipo I/metabolismo , Elongação da Transcrição Genética , Animais , DNA Topoisomerases Tipo I/deficiência , DNA Topoisomerases Tipo II/deficiência , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/metabolismo , Técnicas de Silenciamento de Genes , Impressão Genômica/genética , Humanos , Camundongos , Mutação/genética , Proteínas de Ligação a Poli-ADP-Ribose , RNA Polimerase II/metabolismo , Sinapses/metabolismo , Inibidores da Topoisomerase/farmacologia , Topotecan/farmacologia , Elongação da Transcrição Genética/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...