Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 8(1): 17348, 2018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30478432

RESUMO

The inner ear is a complex structure responsible for hearing and balance, and organ pathology is associated with deafness and balance disorders. To evaluate the role of epigenomic dynamics, we performed whole genome bisulfite sequencing at key time points during the development and maturation of the mouse inner ear sensory epithelium (SE). Our single-nucleotide resolution maps revealed variations in both general characteristics and dynamics of DNA methylation over time. This allowed us to predict the location of non-coding regulatory regions and to identify several novel candidate regulatory factors, such as Bach2, that connect stage-specific regulatory elements to molecular features that drive the development and maturation of the SE. Constructing in silico regulatory networks around sites of differential methylation enabled us to link key inner ear regulators, such as Atoh1 and Stat3, to pathways responsible for cell lineage determination and maturation, such as the Notch pathway. We also discovered that a putative enhancer, defined as a low methylated region (LMR), can upregulate the GJB6 gene and a neighboring non-coding RNA. The study of inner ear SE methylomes revealed novel regulatory regions in the hearing organ, which may improve diagnostic capabilities, and has the potential to guide the development of therapeutics for hearing loss by providing multiple intervention points for manipulation of the auditory system.


Assuntos
Conexina 30/genética , Metilação de DNA/fisiologia , Orelha Interna/embriologia , Orelha Interna/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Surdez/genética , Orelha Interna/citologia , Elementos Facilitadores Genéticos , Epitélio/embriologia , Epitélio/crescimento & desenvolvimento , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Fatores do Domínio POU/genética , Gravidez , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
2.
Hum Mol Genet ; 27(5): 860-874, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29325119

RESUMO

The peripheral deafness gene Mir96 is expressed in both the cochlea and central auditory circuits. To investigate whether it plays a role in the auditory system beyond the cochlea, we characterized homozygous Dmdo/Dmdo mice with a point mutation in miR-96. Anatomical analysis demonstrated a significant decrease in volume of auditory nuclei in Dmdo/Dmdo mice. This decrease resulted from decreased cell size. Non-auditory structures in the brainstem of Dmdo/Dmdo mice or auditory nuclei of the congenital deaf Cldn14-/- mice revealed no such differences. Electrophysiological analysis in the medial nucleus of the trapezoid body (MNTB) showed that principal neurons fired preferentially multiple action potentials upon depolarization, in contrast to the single firing pattern prevalent in controls and Cldn14-/- mice. Immunohistochemistry identified significantly reduced expression of two predicted targets of the mutated miR-96, Kv1.6 and BK channel proteins, possibly contributing to the electrophysiological phenotype. Microscopic analysis of the Dmdo/Dmdo calyx of Held revealed a largely absent compartmentalized morphology, as judged by SV2-labeling. Furthermore, MNTB neurons from Dmdo/Dmdo mice displayed larger synaptic short-term depression, slower AMPA-receptor decay kinetics and a larger NMDA-receptor component, reflecting a less matured stage. Again, these synaptic differences were not present between controls and Cldn14-/- mice. Thus, deafness genes differentially affect the auditory brainstem. Furthermore, our study identifies miR-96 as an essential gene regulatory network element of the auditory system which is required for functional maturation in the peripheral and central auditory system alike.


Assuntos
MicroRNAs/fisiologia , Rombencéfalo/crescimento & desenvolvimento , Rombencéfalo/patologia , Animais , Tamanho Celular , Claudinas/genética , Núcleo Coclear/crescimento & desenvolvimento , Núcleo Coclear/patologia , Regulação da Expressão Gênica no Desenvolvimento , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Camundongos Knockout , Camundongos Mutantes , Mutação , Plasticidade Neuronal , Neurônios/patologia , Superfamília Shaker de Canais de Potássio/genética , Sinapses/patologia , Transmissão Sináptica
3.
Sci Rep ; 7(1): 8637, 2017 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-28819115

RESUMO

Mammalian genomes encode multiple layers of regulation, including a class of RNA molecules known as long non-coding RNAs (lncRNAs). These are >200 nucleotides in length and similar to mRNAs, they are capped, polyadenylated, and spliced. In contrast to mRNAs, lncRNAs are less abundant and have higher tissue specificity, and have been linked to development, epigenetic processes, and disease. However, little is known about lncRNA function in the auditory and vestibular systems, or how they play a role in deafness and vestibular dysfunction. To help address this need, we performed a whole-genome identification of lncRNAs using RNA-seq at two developmental stages of the mouse inner ear sensory epithelium of the cochlea and vestibule. We identified 3,239 lncRNA genes, most of which were intergenic (lincRNAs) and 721 are novel. We examined temporal and tissue specificity by analyzing the developmental profiles on embryonic day 16.5 and at birth. The spatial and temporal patterns of three lncRNAs, two of which are in proximity to genes associated with hearing and deafness, were explored further. Our findings indicate that lncRNAs are prevalent in the sensory epithelium of the mouse inner ear and are likely to play key roles in regulating critical pathways for hearing and balance.


Assuntos
Córtex Auditivo/metabolismo , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , RNA Longo não Codificante/genética , Transcriptoma , Vestíbulo do Labirinto/metabolismo , Animais , Cóclea/metabolismo , Orelha Interna/metabolismo , Regulação da Expressão Gênica , Camundongos
4.
BMC Genomics ; 18(1): 305, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28420336

RESUMO

BACKGROUND: The quantitative relations between RNA and protein are fundamental to biology and are still not fully understood. Across taxa, it was demonstrated that the protein-to-mRNA ratio in steady state varies in a direction that lessens the change in protein levels as a result of changes in the transcript abundance. Evidence for this behavior in tissues is sparse. We tested this phenomenon in new data that we produced for the mouse auditory system, and in previously published tissue datasets. A joint analysis of the transcriptome and proteome was performed across four datasets: inner-ear mouse tissues, mouse organ tissues, lymphoblastoid primate samples and human cancer cell lines. RESULTS: We show that the protein levels are more conserved than the mRNA levels in all datasets, and that changes in transcription are associated with translational changes that exert opposite effects on the final protein level, in all tissues except cancer. Finally, we observe that some functions are enriched in the inner ear on the mRNA level but not in protein. CONCLUSIONS: We suggest that partial buffering between transcription and translation ensures that proteins can be made rapidly in response to a stimulus. Accounting for the buffering can improve the prediction of protein levels from mRNA levels.


Assuntos
Neoplasias/genética , Proteoma/genética , RNA Mensageiro/genética , Transcriptoma/genética , Animais , Proliferação de Células , Orelha Interna/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Especificidade de Órgãos/genética , Primatas/genética , RNA Mensageiro/biossíntese
5.
Mamm Genome ; 27(1-2): 29-46, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26662512

RESUMO

The planar cell polarity (PCP) pathway is responsible for polarizing and orienting cochlear hair cells during development through movement of a primary cilium, the kinocilium. GPSM2/LGN, a mitotic spindle-orienting protein associated with deafness in humans, is a PCP effector involved in kinocilium migration. Here, we link human and mouse truncating mutations in the GPSM2/LGN gene, both leading to hearing loss. The human variant, p.(Trp326*), was identified by targeted genomic enrichment of genes associated with deafness, followed by massively parallel sequencing. Lgn (ΔC) mice, with a targeted deletion truncating the C-terminal GoLoco motifs, are profoundly deaf and show misorientation of the hair bundle and severe malformations in stereocilia shape that deteriorates over time. Full-length protein levels are greatly reduced in mutant mice, with upregulated mRNA levels. The truncated Lgn (ΔC) allele is translated in vitro, suggesting that mutant mice may have partially functioning Lgn. Gαi and aPKC, known to function in the same pathway as Lgn, are dependent on Lgn for proper localization. The polarization of core PCP proteins is not affected in Lgn mutants; however, Lgn and Gαi are misoriented in a PCP mutant, supporting the role of Lgn as a PCP effector. The kinocilium, previously shown to be dependent on Lgn for robust localization, is essential for proper localization of Lgn, as well as Gαi and aPKC, suggesting that cilium function plays a role in positioning of apical proteins. Taken together, our data provide a mechanism for the loss of hearing found in human patients with GPSM2/LGN variants.


Assuntos
Proteínas de Transporte/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Células Ciliadas Auditivas/metabolismo , Perda Auditiva Neurossensorial/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteína Quinase C/genética , Alelos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Movimento Celular , Polaridade Celular , Cílios/genética , Cílios/metabolismo , Cílios/patologia , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Células Ciliadas Auditivas/patologia , Perda Auditiva Neurossensorial/metabolismo , Perda Auditiva Neurossensorial/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Motivos de Nucleotídeos , Linhagem , Proteína Quinase C/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais
6.
Otolaryngol Clin North Am ; 48(6): 1041-61, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26443487

RESUMO

Hearing loss (HL) is one of the most common birth defects in developed countries and is a diverse pathologic condition with different classifications. One of these is based on the association with other clinical features, defined as syndromic hearing loss (SHL). Determining the cause of the HL in these patients is extremely beneficial as it enables a personalized approach to caring for the individual. Early screening can further aid in optimal rehabilitation for a child's development and growth. The advancement of high-throughput sequencing technology is facilitating rapid and low-cost diagnostics for patients with SHL.


Assuntos
Biomarcadores/análise , Diagnóstico Precoce , Bócio Nodular/genética , Perda Auditiva Neurossensorial/genética , Síndromes de Usher/genética , Síndrome de Waardenburg/genética , Criança , Ligação Genética , Bócio Nodular/diagnóstico , Perda Auditiva Neurossensorial/diagnóstico , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mutação , Síndromes de Usher/diagnóstico , Síndrome de Waardenburg/diagnóstico
7.
BMC Genomics ; 15: 484, 2014 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-24942165

RESUMO

BACKGROUND: The mammalian inner ear contains sensory organs, the organ of Corti in the cochlea and cristae and maculae in the vestibule, with each comprised of patterned sensory epithelia that are responsible for hearing and balance. The development, cell fate, patterning, and innervation of both the sensory and nonsensory regions of the inner ear are governed by tight regulation involving, among others, transcription factors and microRNAs (miRNAs). In humans, mutations in specific miRNA genes are associated with hearing loss. In mice, experimental reduction or mutations of miRNAs in the inner ear leads to severe developmental and structural abnormalities. A comprehensive identification of miRNAs in the sensory epithelia and their gene targets will enable pathways of auditory and vestibular function to be defined. RESULTS: In this study, we used Next-Generation Sequencing (NGS) to identify the most prominent miRNAs in the inner ear and to define miRNA-target pairs that form pathways crucial for the function of the sensory epithelial cells. NGS of RNA from inner ear sensory epithelial cells led to the identification of 455 miRNAs in both cochlear and vestibular sensory epithelium, with 30 and 44 miRNAs found in only cochlea or vestibule, respectively. miR-6715-3p and miR-6715-5p were defined for the first time in the inner ear. Gene targets were identified for each of these miRNAs, including Arhgap12, a GTPase activating protein, for miR-6715-3p, implicating this miRNA in sensory hair cell bundle development, actin reorganization, cell adhesion and inner ear morphogenesis. CONCLUSIONS: This study provides a comprehensive atlas of miRNAs in the inner ear sensory epithelia. The results provide further support of the essential regulatory role of miRNAs in inner ear sensory epithelia and in regulating pathways that define development and growth of these cells.


Assuntos
Orelha Interna/metabolismo , Epitélio/metabolismo , MicroRNAs/genética , Transdução de Sinais , Animais , Cóclea/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Interferência de RNA , RNA Mensageiro/genética , Transcriptoma , Vestíbulo do Labirinto/metabolismo
8.
J Biol Chem ; 288(36): 25865-25879, 2013 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-23893414

RESUMO

The superior olivary complex (SOC) is an essential auditory brainstem relay involved in sound localization. To identify the genetic program underlying its maturation, we profiled the rat SOC transcriptome at postnatal days 0, 4, 16, and 25 (P0, P4, P16, and P25, respectively), using genome-wide microarrays (41,012 oligonucleotides (oligos)). Differences in gene expression between two consecutive stages were highest between P4 and P16 (3.6%) and dropped to 0.06% between P16 and P25. To identify SOC-related genetic programs, we also profiled the entire brain at P4 and P25. The number of differentially expressed oligonucleotides between SOC and brain almost doubled from P4 to P25 (4.4% versus 7.6%). These data demonstrate considerable molecular specification around hearing onset, which is rapidly finalized. Prior to hearing onset, several transcription factors associated with the peripheral auditory system were up-regulated, probably coordinating the development of the auditory system. Additionally, crystallin-γ subunits and serotonin-related genes were highly expressed. The molecular repertoire of mature neurons was sculpted by SOC-related up- and down-regulation of voltage-gated channels and G-proteins. Comparison with the brain revealed a significant enrichment of hearing impairment-related oligos in the SOC (26 in the SOC, only 11 in the brain). Furthermore, 29 of 453 SOC-related oligos mapped within 19 genetic intervals associated with hearing impairment. Together, we identified sequential genetic programs in the SOC, thereby pinpointing candidates that may guide its development and ensure proper function. The enrichment of hearing impairment-related genes in the SOC may have implications for restoring hearing because central auditory structures might be more severely affected than previously appreciated.


Assuntos
Tronco Encefálico , Regulação da Expressão Gênica/fisiologia , Audição/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Transcriptoma/fisiologia , Animais , Animais Recém-Nascidos , Tronco Encefálico/citologia , Tronco Encefálico/crescimento & desenvolvimento , Tronco Encefálico/metabolismo , Feminino , Humanos , Masculino , Proteínas do Tecido Nervoso/genética , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Sprague-Dawley
9.
Hum Mutat ; 34(8): 1102-10, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23606368

RESUMO

POU3F4 is a POU domain transcription factor that is required for hearing. In the ear, POU3F4 is essential for mesenchymal remodeling of the bony labyrinth and is the causative gene for DFNX2 human nonsyndromic deafness. Ear abnormalities underlie this form of deafness, characterized previously in multiple spontaneous, radiation-induced and transgenic mouse mutants. Here, we report three novel mutations in the POU3F4 gene that result in profound hearing loss in both humans and mice. A p.Gln79* mutation was identified in a child from an Israeli family, revealed by massively parallel sequencing (MPS). This strategy demonstrates the strength of MPS for diagnosis with only one affected individual. A second mutation, p.Ile285Argfs*43, was identified by Sanger sequencing. A p.Cys300* mutation was found in an ENU-induced mutant mouse, schwindel (sdl), by positional cloning. The mutation leads to a predicted truncated protein, similar to the human mutations, providing a relevant mouse model. The p.Ile285Argfs*43 and p.Cys300* mutations lead to a shift of Pou3f4 nuclear localization to the cytoplasm, demonstrated in cellular localization studies and in the inner ears of the mutant mice. The discovery of these mutations facilitates a deeper comprehension of the molecular basis of inner ear defects due to mutations in the POU3F4 transcription factor.


Assuntos
Citoplasma/metabolismo , Surdez/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fatores do Domínio POU/genética , Fatores do Domínio POU/metabolismo , Animais , Células COS , Núcleo Celular/metabolismo , Criança , Chlorocebus aethiops , Surdez/metabolismo , Orelha Interna/metabolismo , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
10.
Front Mol Neurosci ; 6: 52, 2013 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-24391537

RESUMO

Non-coding microRNAs (miRNAs) have a fundamental role in gene regulation and expression in almost every multicellular organism. Only discovered in the last decade, miRNAs are already known to play a leading role in many aspects of disease. In the vertebrate inner ear, miRNAs are essential for controlling development and survival of hair cells. Moreover, dysregulation of miRNAs has been implicated in sensorineural hearing impairment, as well as in other ear diseases such as cholesteatomas, vestibular schwannomas, and otitis media. Due to the inaccessibility of the ear in humans, animal models have provided the optimal tools to study miRNA expression and function, in particular mice and zebrafish. A major focus of current research has been to discover the targets of the miRNAs expressed in the inner ear, in order to determine the regulatory pathways of the auditory and vestibular systems. The potential for miRNAs manipulation in development of therapeutic tools for hearing impairment is as yet unexplored, paving the way for future work in the field.

11.
Biochim Biophys Acta ; 1813(12): 2000-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21878356

RESUMO

Recent evidence suggests that coupled leading and lagging strand DNA synthesis operates in mammalian mitochondrial DNA (mtDNA) replication, but the factors involved in lagging strand synthesis are largely uncharacterised. We investigated the effect of knockdown of the candidate proteins in cultured human cells under conditions where mtDNA appears to replicate chiefly via coupled leading and lagging strand DNA synthesis to restore the copy number of mtDNA to normal levels after transient mtDNA depletion. DNA ligase III knockdown attenuated the recovery of mtDNA copy number and appeared to cause single strand nicks in replicating mtDNA molecules, suggesting the involvement of DNA ligase III in Okazaki fragment ligation in human mitochondria. Knockdown of ribonuclease (RNase) H1 completely prevented the mtDNA copy number restoration, and replication intermediates with increased single strand nicks were readily observed. On the other hand, knockdown of neither flap endonuclease 1 (FEN1) nor DNA2 affected mtDNA replication. These findings imply that RNase H1 is indispensable for the progression of mtDNA synthesis through removing RNA primers from Okazaki fragments. In the nucleus, Okazaki fragments are ligated by DNA ligase I, and the RNase H2 is involved in Okazaki fragment processing. This study thus proposes that the mitochondrial replication system utilises distinct proteins, DNA ligase III and RNase H1, for Okazaki fragment maturation.


Assuntos
Neoplasias Ósseas/genética , DNA Ligases/metabolismo , Replicação do DNA , DNA Mitocondrial/genética , DNA/metabolismo , Osteossarcoma/genética , Ribonuclease H/metabolismo , Southern Blotting , Western Blotting , Neoplasias Ósseas/metabolismo , DNA/genética , DNA Ligase Dependente de ATP , DNA Ligases/genética , Endonucleases Flap/genética , Endonucleases Flap/metabolismo , Humanos , Osteossarcoma/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose , Ribonuclease H/genética , Timidina Quinase/deficiência , Células Tumorais Cultivadas , Proteínas de Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...