Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 126(11): 4319-4330, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27721240

RESUMO

Huntington's disease (HD) is a polyglutamine disorder caused by a CAG expansion in the Huntingtin (HTT) gene exon 1. This expansion encodes a mutant protein whose abnormal function is traditionally associated with HD pathogenesis; however, recent evidence has also linked HD pathogenesis to RNA stable hairpins formed by the mutant HTT expansion. Here, we have shown that a locked nucleic acid-modified antisense oligonucleotide complementary to the CAG repeat (LNA-CTG) preferentially binds to mutant HTT without affecting HTT mRNA or protein levels. LNA-CTGs produced rapid and sustained improvement of motor deficits in an R6/2 mouse HD model that was paralleled by persistent binding of LNA-CTG to the expanded HTT exon 1 transgene. Motor improvement was accompanied by a pronounced recovery in the levels of several striatal neuronal markers severely impaired in R6/2 mice. Furthermore, in R6/2 mice, LNA-CTG blocked several pathogenic mechanisms caused by expanded CAG RNA, including small RNA toxicity and decreased Rn45s expression levels. These results suggest that LNA-CTGs promote neuroprotection by blocking the detrimental activity of CAG repeats within HTT mRNA. The present data emphasize the relevance of expanded CAG RNA to HD pathogenesis, indicate that inhibition of HTT expression is not required to reverse motor deficits, and further suggest a therapeutic potential for LNA-CTG in polyglutamine disorders.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Huntingtina , Doença de Huntington , RNA Antissenso , Repetições de Trinucleotídeos , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Proteína Huntingtina/biossíntese , Proteína Huntingtina/genética , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/terapia , Masculino , Camundongos , Camundongos Transgênicos , RNA Antissenso/genética , RNA Antissenso/farmacologia
2.
Genome Biol ; 15(4): R57, 2014 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-24708865

RESUMO

BACKGROUND: MicroRNAs (miRNAs) are established regulators of development, cell identity and disease. Although nearly two thousand human miRNA genes are known and new ones are continuously discovered, no attempt has been made to gauge the total miRNA content of the human genome. RESULTS: Employing an innovative computational method on massively pooled small RNA sequencing data, we report 2,469 novel human miRNA candidates of which 1,098 are validated by in-house and published experiments. Almost 300 candidates are robustly expressed in a neuronal cell system and are regulated during differentiation or when biogenesis factors Dicer, Drosha, DGCR8 or Ago2 are silenced. To improve expression profiling, we devised a quantitative miRNA capture system. In a kidney cell system, 400 candidates interact with DGCR8 at transcript positions that suggest miRNA hairpin recognition, and 1,000 of the new miRNA candidates interact with Ago1 or Ago2, indicating that they are directly bound by miRNA effector proteins. From kidney cell CLASH experiments, in which miRNA-target pairs are ligated and sequenced, we observe hundreds of interactions between novel miRNAs and mRNA targets. The novel miRNA candidates are specifically but lowly expressed, raising the possibility that not all may be functional. Interestingly, the majority are evolutionarily young and overrepresented in the human brain. CONCLUSIONS: In summary, we present evidence that the complement of human miRNA genes is substantially larger than anticipated, and that more are likely to be discovered in the future as more tissues and experimental conditions are sequenced to greater depth.


Assuntos
Genoma Humano , MicroRNAs/genética , Proteínas Argonautas/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Humanos , Células MCF-7 , MicroRNAs/metabolismo , Ligação Proteica , Proteínas de Ligação a RNA/metabolismo
3.
RNA Biol ; 10(7): 1093-106, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23673382

RESUMO

MicroRNAs (miRNAs) and other small non-coding RNAs (sncRNAs) are post-transcriptional regulators of gene expression, playing key roles in neuronal development, plasticity, and disease. Transcriptome deregulation caused by miRNA dysfunction has been associated to neurodegenerative diseases. Parkinson disease (PD) is the second most common neurodegenerative disease showing deregulation of the coding and small non-coding transcriptome. On profiling sncRNA in PD brain areas differently affected, we found that upregulation of a small vault RNA (svtRNA2-1a) is widespread in PD brains, occurring early in the course of the disease (at pre-motor stages). SvtRNA2-1a biogenesis was dependent on Dicer activity on its precursor (vtRNA2-1) but independent of Drosha endonuclease, unlike the canonical miRNAs. Although endogenous svtRNA2-1a was enriched in Ago-2 immunoprecipitates in differentiated SH-SY5Y neuronal cells, overexpression of svtRNA2-1a induced subtle transcriptomic changes, suggesting that gene expression regulation may involve other mechanisms than mRNA decay only. Function enrichment analysis of the genes deregulated by svtRNA2-1a overexpression or svtRNA2-1a predicted targets identified pathways related to nervous system development and cell type specification. The expression pattern of svtRNA2-1a during development and aging of the human brain and the detrimental consequences of a svtRNA2-1a mimic overexpression in neuronal cells further indicate that low svtRNA2-1a levels may be important for the maintenance of neurons. Our results suggest that early svtRNA2-1a upregulation in PD may contribute to perturbations of gene expression networks, underlying metabolic impairment and cell dysfunction. A better understanding of the pathways regulated by svtRNA2-a, and also the mechanisms regulating its expression should facilitate the identification of new targets for therapeutic approaches in PD.


Assuntos
Regulação da Expressão Gênica , Neurônios/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Pequeno RNA não Traduzido/genética , Proteínas Argonautas/metabolismo , Sequência de Bases , Encéfalo/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Expressão Gênica , Humanos , Dados de Sequência Molecular , Neurônios/citologia , Conformação de Ácido Nucleico , Fases de Leitura Aberta , Doença de Parkinson/tratamento farmacológico , Pequeno RNA não Traduzido/química , Reprodutibilidade dos Testes , Ribonuclease III/metabolismo , Alinhamento de Sequência , Transcriptoma , Regulação para Cima
4.
PLoS Genet ; 8(2): e1002481, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22383888

RESUMO

Huntington's disease (HD) is an autosomal dominantly inherited disorder caused by the expansion of CAG repeats in the Huntingtin (HTT) gene. The abnormally extended polyglutamine in the HTT protein encoded by the CAG repeats has toxic effects. Here, we provide evidence to support that the mutant HTT CAG repeats interfere with cell viability at the RNA level. In human neuronal cells, expanded HTT exon-1 mRNA with CAG repeat lengths above the threshold for complete penetrance (40 or greater) induced cell death and increased levels of small CAG-repeated RNAs (sCAGs), of ≈21 nucleotides in a Dicer-dependent manner. The severity of the toxic effect of HTT mRNA and sCAG generation correlated with CAG expansion length. Small RNAs obtained from cells expressing mutant HTT and from HD human brains significantly decreased neuronal viability, in an Ago2-dependent mechanism. In both cases, the use of anti-miRs specific for sCAGs efficiently blocked the toxic effect, supporting a key role of sCAGs in HTT-mediated toxicity. Luciferase-reporter assays showed that expanded HTT silences the expression of CTG-containing genes that are down-regulated in HD. These results suggest a possible link between HD and sCAG expression with an aberrant activation of the siRNA/miRNA gene silencing machinery, which may trigger a detrimental response. The identification of the specific cellular processes affected by sCAGs may provide insights into the pathogenic mechanisms underlying HD, offering opportunities to develop new therapeutic approaches.


Assuntos
Doença de Huntington/etiologia , Proteínas do Tecido Nervoso/genética , RNA Mensageiro , Éxons , Inativação Gênica , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/patologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Peptídeos/genética , Peptídeos/metabolismo , RNA Mensageiro/metabolismo , Expansão das Repetições de Trinucleotídeos
5.
Hum Mol Genet ; 20(15): 3067-78, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21558425

RESUMO

MicroRNAs (miRNAs) are post-transcriptional gene expression regulators, playing key roles in neuronal development, plasticity and disease. Parkinson's disease (PD) is the second most common neurodegenerative disorder, characterized by the presence of protein inclusions or Lewy bodies and a progressive loss of dopaminergic neurons in the midbrain. Here, we have evaluated miRNA expression deregulation in PD brain samples. MiRNA expression profiling revealed decreased expression of miR-34b and miR-34c in brain areas with variable neuropathological affectation at clinical (motor) stages (Braak stages 4 and 5) of the disease, including the amygdala, frontal cortex, substantia nigra and cerebellum. Furthermore, misregulation of miR-34b/c was detected in pre-motor stages (stages 1-3) of the disease, and thus in cases that did not receive any PD-related treatment during life. Depletion of miR-34b or miR-34c in differentiated SH-SY5Y dopaminergic neuronal cells resulted in a moderate reduction in cell viability that was accompanied by altered mitochondrial function and dynamics, oxidative stress and reduction in total cellular adenosin triphosphate content. MiR-34b/c downregulation was coupled to a decrease in the expression of DJ1 and Parkin, two proteins associated to familial forms of PD that also have a role in idiopathic cases. Accordingly, DJ1 and Parkin expression was reduced in PD brain samples displaying strong miR-34b/c downregulation. We propose that early deregulation of miR-34b/c in PD triggers downstream transcriptome alterations underlying mitochondrial dysfunction and oxidative stress, which ultimately compromise cell viability. A better understanding of the cellular pathways controlling and/or controlled by miR-34b/c should allow identification of targets for development of therapeutic approaches.


Assuntos
MicroRNAs/genética , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Western Blotting , Linhagem Celular Tumoral , Humanos , Microscopia Eletrônica de Transmissão , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/ultraestrutura , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real
6.
Biol Psychiatry ; 69(6): 526-33, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21168126

RESUMO

BACKGROUND: The involvement of microRNAs (miRNAs) in neuronal differentiation and synaptic plasticity suggests a role for miRNAs in psychiatric disorders; association analyses and functional approaches were used to evaluate the implication of miRNAs in the susceptibility for panic disorder. METHODS: Case-control studies for 712 single-nucleotide polymorphisms (SNPs) tagging 325 human miRNA regions were performed in 203 Spanish patients with panic disorder and 341 control subjects. A sample of 321 anxiety patients and 642 control subjects from Finland and 102 panic disorder patients and 829 control subjects from Estonia was used as a replica. Reporter-gene assays and miRNA overexpression experiments in neuroblastoma cells were used to functionally evaluate the spectrum of genes regulated by the associated miRNAs. RESULTS: Two SNPs associated with panic disorder: rs6502892 tagging miR-22 (p < .0002), and rs11763020 tagging miR-339 (p < .00008). Other SNPs tagging miR-138-2, miR-488, miR-491, and miR-148a regions associated with different panic disorder phenotypes. Replication in the north-European sample supported several of these associations, although they did not pass correction for multiple testing. Functional studies revealed that miR-138-2, miR-148a, and miR-488 repress (30%-60%) several candidate genes for panic disorder--GABRA6, CCKBR and POMC, respectively--and that miR-22 regulates four other candidate genes: BDNF, HTR2C, MAOA, and RGS2. Transcriptome analysis of neuroblastoma cells transfected with miR-22 and miR-488 showed altered expression of a subset of predicted target genes for these miRNAs and of genes that might be affecting physiological pathways related to anxiety. CONCLUSIONS: This work represents the first report of a possible implication of miRNAs in the etiology of panic disorder.


Assuntos
Ansiedade/genética , Predisposição Genética para Doença , MicroRNAs/genética , Transtorno de Pânico/genética , Polimorfismo de Nucleotídeo Único/genética , Adulto , Estudos de Casos e Controles , Linhagem Celular Tumoral , Comparação Transcultural , Estônia , Feminino , Finlândia , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/genética , Estudo de Associação Genômica Ampla/métodos , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Monoaminoxidase/genética , Monoaminoxidase/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroblastoma/patologia , Transtorno de Pânico/etnologia , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Receptores da Colecistocinina/genética , Receptores da Colecistocinina/metabolismo , Receptores de GABA-A/metabolismo , Receptores 5-HT2 de Serotonina/genética , Receptores 5-HT2 de Serotonina/metabolismo , Espanha , Transfecção , Adulto Jovem
7.
BMC Mol Biol ; 11: 95, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21143953

RESUMO

BACKGROUND: Neurotrophins and their receptors are key molecules in the regulation of neuronal differentiation and survival. They mediate the survival of neurons during development and adulthood and are implicated in synaptic plasticity. The human neurotrophin-3 receptor gene NTRK3 yields two major isoforms, a full-length kinase-active form and a truncated non-catalytic form, which activates a specific pathway affecting membrane remodeling and cytoskeletal reorganization. The two variants present non-overlapping 3'UTRs, indicating that they might be differentially regulated at the post-transcriptional level. Here, we provide evidence that the two isoforms of NTRK3 are targeted by different sets of microRNAs, small non-coding RNAs that play an important regulatory role in the nervous system. RESULTS: We identify one microRNA (miR-151-3p) that represses the full-length isoform of NTRK3 and four microRNAs (miR-128, miR-485-3p, miR-765 and miR-768-5p) that repress the truncated isoform. In particular, we show that the overexpression of miR-128 - a brain enriched miRNA - causes morphological changes in SH-SY5Y neuroblastoma cells similar to those observed using an siRNA specifically directed against truncated NTRK3, as well as a significant increase in cell number. Accordingly, transcriptome analysis of cells transfected with miR-128 revealed an alteration of the expression of genes implicated in cytoskeletal organization as well as genes involved in apoptosis, cell survival and proliferation, including the anti-apoptotic factor BCL2. CONCLUSIONS: Our results show that the regulation of NTRK3 by microRNAs is isoform-specific and suggest that neurotrophin-mediated processes are strongly linked to microRNA-dependent mechanisms. In addition, these findings open new perspectives for the study of the physiological role of miR-128 and its possible involvement in cell death/survival processes.


Assuntos
MicroRNAs/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptor trkC/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Biologia Computacional , Perfilação da Expressão Gênica , Humanos , MicroRNAs/genética , Fatores de Crescimento Neural/metabolismo , Neuroblastoma , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/metabolismo , Receptor trkC/antagonistas & inibidores , Regulação para Cima
8.
Hum Mol Genet ; 19(20): 4017-25, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20656788

RESUMO

Previous studies in mice have reported five different microRNAs (miRNAs; miR-219-1/132/183/96/182) to be modulators of the endogenous circadian clock and have presented experimental evidence for some of the genes involved in the molecular clock machinery as target sites. Moreover, disruption of circadian rhythms has long been implicated in the pathophysiology of major depression (MD). We investigated these miRNAs and some of their target sites at the sequence and functional levels as possible predisposing factors for susceptibility to MD and related chronobiological subphenotypes. Mutational screening was performed in a sample of 359 MD patients and 341 control individuals. We found a significant association between the T allele of the rs76481776 polymorphism in the pre-miR-182 and late insomnia in MD patients. Previous studies have reported an association between insomnia and CLOCK gene, a predicted miR-182 target site. A significant overexpression of miR-182 was detected by quantitative real-time polymerase chain reaction in cells transfected with the mutated form of the pre-miR-182 when compared with wild-type form. Moreover, a significant reduction in luciferase activity of plasmids with 3' UTR of ADCY6, CLOCK and DSIP genes was shown when transfecting cells with the mutated form of pre-miR-182 compared with cells that did not express miR-182. These data indicate that abnormal processing of pre-miR-182 in patients carrying the T allele of the rs76481776 polymorphism may contribute to the dysregulation of circadian rhythms in MD patients with insomnia, which could influence expression levels of the mature form of miR-182 and might increase downregulation in some of its target genes.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/genética , Transtorno Depressivo Maior/genética , Transtorno Depressivo Maior/fisiopatologia , MicroRNAs/genética , Distúrbios do Início e da Manutenção do Sono/genética , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Análise Mutacional de DNA , Regulação da Expressão Gênica , Predisposição Genética para Doença , Variação Genética , Vetores Genéticos , Humanos , MicroRNAs/metabolismo , Plasmídeos , Reação em Cadeia da Polimerase , Polimorfismo Genético , Precursores de RNA/genética , Precursores de RNA/metabolismo , Alinhamento de Sequência , Distúrbios do Início e da Manutenção do Sono/fisiopatologia , Transfecção
9.
Hum Mutat ; 30(7): 1062-71, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19370765

RESUMO

Genetic and functional data indicate that variation in the expression of the neurotrophin-3 receptor gene (NTRK3) may have an impact on neuronal plasticity, suggesting a role for NTRK3 in the pathophysiology of anxiety disorders. MicroRNA (miRNA) posttranscriptional gene regulators act by base-pairing to specific sequence sites, usually at the 3'UTR of the target mRNA. Variants at these sites might result in gene expression changes contributing to disease susceptibility. We investigated genetic variation in two different isoforms of NTRK3 as candidate susceptibility factors for anxiety by resequencing their 3'UTRs in patients with panic disorder (PD), obsessive-compulsive disorder (OCD), and in controls. We have found the C allele of rs28521337, located in a functional target site for miR-485-3p in the truncated isoform of NTRK3, to be significantly associated with the hoarding phenotype of OCD. We have also identified two new rare variants in the 3'UTR of NTRK3, ss102661458 and ss102661460, each present only in one chromosome of a patient with PD. The ss102661458 variant is located in a functional target site for miR-765, and the ss102661460 in functional target sites for two miRNAs, miR-509 and miR-128, the latter being a brain-enriched miRNA involved in neuronal differentiation and synaptic processing. Interestingly, these two variants significantly alter the miRNA-mediated regulation of NTRK3, resulting in recovery of gene expression. These data implicate miRNAs as key posttranscriptional regulators of NTRK3 and provide a framework for allele-specific miRNA regulation of NTRK3 in anxiety disorders.


Assuntos
Alelos , Transtornos de Ansiedade/genética , Predisposição Genética para Doença , Variação Genética , Receptor trkC/genética , Regiões 3' não Traduzidas , Adolescente , Adulto , Estudos de Casos e Controles , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Transtorno Obsessivo-Compulsivo/genética , Transtorno de Pânico/genética , Isoformas de Proteínas/genética , Adulto Jovem
10.
Immunogenetics ; 55(2): 116-21, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12709835

RESUMO

SH2D1A, the X-linked lymphoproliferative disease (XLP) gene, encodes a cytoplasmic protein that plays an essential role in controlling Epstein-Barr virus infection. It is expressed in T and NK cells, but not in B cells or in granulocytes. The promoter, the regulatory regions, as well as the mechanisms controlling its tissue-specific expression, are still unknown. We tested the hypothesis that DNA methylation might contribute to tissue-specific SH2D1A gene expression and analyzed the methylation status of 2,300 bp upstream of the ATG starting codon, the coding region and part of intron 1. By bisulfite sequencing and methylation-sensitive restriction enzyme digestion, we show that a differential methylation pattern of CpG-rich regions in the 5' region and the adjacent exon 1 of the SH2D1A gene indeed correlates with the tissue-specific gene transcription.


Assuntos
Proteínas de Transporte/genética , Metilação de DNA , Peptídeos e Proteínas de Sinalização Intracelular , Transtornos Linfoproliferativos/genética , Animais , Sítios de Ligação , Biologia Computacional , Ilhas de CpG , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Análise de Sequência de DNA , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA