Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27.053
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 174(5): 1229-1246.e17, 2018 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-30078709

RESUMO

In the auditory system, type I spiral ganglion neurons (SGNs) convey complex acoustic information from inner hair cells (IHCs) to the brainstem. Although SGNs exhibit variation in physiological and anatomical properties, it is unclear which features are endogenous and which reflect input from synaptic partners. Using single-cell RNA sequencing, we derived a molecular classification of mouse type I SGNs comprising three subtypes that express unique combinations of Ca2+ binding proteins, ion channel regulators, guidance molecules, and transcription factors. Based on connectivity and susceptibility to age-related loss, these subtypes correspond to those defined physiologically. Additional intrinsic differences among subtypes and across the tonotopic axis highlight an unexpectedly active role for SGNs in auditory processing. SGN identities emerge postnatally and are disrupted in a mouse model of deafness that lacks IHC-driven activity. These results elucidate the range, nature, and origins of SGN diversity, with implications for treatment of congenital deafness.


Assuntos
Orelha Interna/fisiologia , Células Ciliadas Auditivas Internas/fisiologia , Células Receptoras Sensoriais/fisiologia , Sistemas de Transporte de Aminoácidos Acídicos/genética , Animais , Calbindina 2/genética , Cóclea/fisiologia , Surdez/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência de RNA , Gânglio Espiral da Cóclea/fisiologia , Transmissão Sináptica , Transgenes
2.
Cell ; 174(3): 536-548.e21, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-29961578

RESUMO

The DNA-binding protein REST forms complexes with histone deacetylases (HDACs) to repress neuronal genes in non-neuronal cells. In differentiating neurons, REST is downregulated predominantly by transcriptional silencing. Here we report that post-transcriptional inactivation of REST by alternative splicing is required for hearing in humans and mice. We show that, in the mechanosensory hair cells of the mouse ear, regulated alternative splicing of a frameshift-causing exon into the Rest mRNA is essential for the derepression of many neuronal genes. Heterozygous deletion of this alternative exon of mouse Rest causes hair cell degeneration and deafness, and the HDAC inhibitor SAHA (Vorinostat) rescues the hearing of these mice. In humans, inhibition of the frameshifting splicing event by a novel REST variant is associated with dominantly inherited deafness. Our data reveal the necessity for alternative splicing-dependent regulation of REST in hair cells, and they identify a potential treatment for a group of hereditary deafness cases.


Assuntos
Surdez/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Processamento Alternativo/genética , Animais , Linhagem Celular , Éxons , Regulação da Expressão Gênica/genética , Células HEK293 , Células Ciliadas Auditivas/fisiologia , Audição/genética , Audição/fisiologia , Inibidores de Histona Desacetilases/metabolismo , Histona Desacetilases/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neurônios , Splicing de RNA/genética , Proteínas Repressoras/fisiologia , Fatores de Transcrição , Vorinostat/farmacologia
3.
Nat Rev Genet ; 24(10): 665-686, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37173518

RESUMO

Progress in deciphering the genetic architecture of human sensorineural hearing impairment (SNHI) or loss, and multidisciplinary studies of mouse models, have led to the elucidation of the molecular mechanisms underlying auditory system function, primarily in the cochlea, the mammalian hearing organ. These studies have provided unparalleled insights into the pathophysiological processes involved in SNHI, paving the way for the development of inner-ear gene therapy based on gene replacement, gene augmentation or gene editing. The application of these approaches in preclinical studies over the past decade has highlighted key translational opportunities and challenges for achieving effective, safe and sustained inner-ear gene therapy to prevent or cure monogenic forms of SNHI and associated balance disorders.


Assuntos
Surdez , Perda Auditiva Neurossensorial , Camundongos , Animais , Humanos , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/terapia , Audição/genética , Terapia Genética , Edição de Genes , Surdez/genética , Surdez/terapia , Mamíferos/genética
4.
Nature ; 613(7943): 317-323, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36544024

RESUMO

Cochlear implants (CIs) are neuroprosthetic devices that can provide hearing to deaf people1. Despite the benefits offered by CIs, the time taken for hearing to be restored and perceptual accuracy after long-term CI use remain highly variable2,3. CI use is believed to require neuroplasticity in the central auditory system, and differential engagement of neuroplastic mechanisms might contribute to the variability in outcomes4-7. Despite extensive studies on how CIs activate the auditory system4,8-12, the understanding of CI-related neuroplasticity remains limited. One potent factor enabling plasticity is the neuromodulator noradrenaline from the brainstem locus coeruleus (LC). Here we examine behavioural responses and neural activity in LC and auditory cortex of deafened rats fitted with multi-channel CIs. The rats were trained on a reward-based auditory task, and showed considerable individual differences of learning rates and maximum performance. LC photometry predicted when CI subjects began responding to sounds and longer-term perceptual accuracy. Optogenetic LC stimulation produced faster learning and higher long-term accuracy. Auditory cortical responses to CI stimulation reflected behavioural performance, with enhanced responses to rewarded stimuli and decreased distinction between unrewarded stimuli. Adequate engagement of central neuromodulatory systems is thus a potential clinically relevant target for optimizing neuroprosthetic device use.


Assuntos
Implantes Cocleares , Surdez , Locus Cerúleo , Animais , Ratos , Implante Coclear , Surdez/fisiopatologia , Surdez/terapia , Audição/fisiologia , Aprendizagem/fisiologia , Locus Cerúleo/citologia , Locus Cerúleo/fisiologia , Plasticidade Neuronal , Norepinefrina/metabolismo , Córtex Auditivo/citologia , Córtex Auditivo/fisiologia , Córtex Auditivo/fisiopatologia , Neurônios/fisiologia , Recompensa , Optogenética , Fotometria
5.
Cell ; 154(6): 1175-7, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-24034237

RESUMO

For their work on the development of the modern cochlear implant, which bestows hearing to individuals with profound deafness, Ingeborg Hochmair, Graeme Clark, and Blake Wilson are the 2013 recipients of the Lasker∼DeBakey Clinical Medical Research Award.


Assuntos
Distinções e Prêmios , Implantes Cocleares/história , Surdez/cirurgia , Implante Coclear , Nervo Coclear/cirurgia , História do Século XX , Humanos , Percepção da Fala , Estados Unidos
6.
Annu Rev Neurosci ; 42: 47-65, 2019 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-30699049

RESUMO

The modern cochlear implant (CI) is the most successful neural prosthesis developed to date. CIs provide hearing to the profoundly hearing impaired and allow the acquisition of spoken language in children born deaf. Results from studies enabled by the CI have provided new insights into (a) minimal representations at the periphery for speech reception, (b) brain mechanisms for decoding speech presented in quiet and in acoustically adverse conditions, (c) the developmental neuroscience of language and hearing, and (d) the mechanisms and time courses of intramodal and cross-modal plasticity. Additionally, the results have underscored the interconnectedness of brain functions and the importance of top-down processes in perception and learning. The findings are described in this review with emphasis on the developing brain and the acquisition of hearing and spoken language.


Assuntos
Percepção Auditiva/fisiologia , Implantes Cocleares , Período Crítico Psicológico , Desenvolvimento da Linguagem , Animais , Transtornos da Percepção Auditiva/etiologia , Encéfalo/crescimento & desenvolvimento , Implante Coclear , Compreensão , Sinais (Psicologia) , Surdez/congênito , Surdez/fisiopatologia , Surdez/psicologia , Surdez/cirurgia , Desenho de Equipamento , Humanos , Transtornos do Desenvolvimento da Linguagem/etiologia , Transtornos do Desenvolvimento da Linguagem/prevenção & controle , Aprendizagem/fisiologia , Plasticidade Neuronal , Estimulação Luminosa
7.
Cell ; 148(4): 716-26, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22341444

RESUMO

Mitochondrial dysfunction causes poorly understood tissue-specific pathology stemming from primary defects in respiration, coupled with altered reactive oxygen species (ROS), metabolic signaling, and apoptosis. The A1555G mtDNA mutation that causes maternally inherited deafness disrupts mitochondrial ribosome function, in part, via increased methylation of the mitochondrial 12S rRNA by the methyltransferase mtTFB1. In patient-derived A1555G cells, we show that 12S rRNA hypermethylation causes ROS-dependent activation of AMP kinase and the proapoptotic nuclear transcription factor E2F1. This retrograde mitochondrial-stress relay is operative in vivo, as transgenic-mtTFB1 mice exhibit enhanced 12S rRNA methylation in multiple tissues, increased E2F1 and apoptosis in the stria vascularis and spiral ganglion neurons of the inner ear, and progressive E2F1-dependent hearing loss. This mouse mitochondrial disease model provides a robust platform for deciphering the complex tissue specificity of human mitochondrial-based disorders, as well as the precise pathogenic mechanism of maternally inherited deafness and its exacerbation by environmental factors.


Assuntos
Surdez/metabolismo , Modelos Animais de Doenças , Fator de Transcrição E2F1/metabolismo , Animais , DNA Mitocondrial/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Orelha Interna/patologia , Cistos Glanglionares/patologia , Humanos , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Mutação , Neurônios/patologia , RNA Ribossômico/metabolismo , Espécies Reativas de Oxigênio , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Proc Natl Acad Sci U S A ; 121(5): e2304680121, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38266052

RESUMO

Mechanosensory hair cells of the mature mammalian organ of Corti do not regenerate; consequently, loss of hair cells leads to permanent hearing loss. Although nonmammalian vertebrates can regenerate hair cells from neighboring supporting cells, many humans with severe hearing loss lack both hair cells and supporting cells, with the organ of Corti being replaced by a flat epithelium of nonsensory cells. To determine whether the mature cochlea can produce hair cells in vivo, we reprogrammed nonsensory cells adjacent to the organ of Corti with three hair cell transcription factors: Gfi1, Atoh1, and Pou4f3. We generated numerous hair cell-like cells in nonsensory regions of the cochlea and new hair cells continued to be added over a period of 9 wk. Significantly, cells adjacent to reprogrammed hair cells expressed markers of supporting cells, suggesting that transcription factor reprogramming of nonsensory cochlear cells in adult animals can generate mosaics of sensory cells like those seen in the organ of Corti. Generating such sensory mosaics by reprogramming may represent a potential strategy for hearing restoration in humans.


Assuntos
Surdez , Células Ciliadas Auditivas , Adulto , Animais , Humanos , Fatores de Transcrição/genética , Epitélio , Cóclea , Mamíferos
9.
Proc Natl Acad Sci U S A ; 121(10): e2309656121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38408254

RESUMO

Inner ear hair cells are characterized by the F-actin-based stereocilia that are arranged into a staircase-like pattern on the apical surface of each hair cell. The tips of shorter-row stereocilia are connected with the shafts of their neighboring taller-row stereocilia through extracellular links named tip links, which gate mechano-electrical transduction (MET) channels in hair cells. Cadherin 23 (CDH23) forms the upper part of tip links, and its cytoplasmic tail is inserted into the so-called upper tip-link density (UTLD) that contains other proteins such as harmonin. The Cdh23 gene is composed of 69 exons, and we show here that exon 68 is subjected to hair cell-specific alternative splicing. Tip-link formation is not affected in genetically modified mutant mice lacking Cdh23 exon 68. Instead, the stability of tip links is compromised in the mutants, which also suffer from progressive and noise-induced hearing loss. Moreover, we show that the cytoplasmic tail of CDH23(+68) but not CDH23(-68) cooperates with harmonin in phase separation-mediated condensate formation. In conclusion, our work provides evidence that inclusion of Cdh23 exon 68 is critical for the stability of tip links through regulating condensate formation of UTLD components.


Assuntos
Surdez , Perda Auditiva , Camundongos , Animais , Perda Auditiva/genética , Perda Auditiva/metabolismo , Células Ciliadas Auditivas/fisiologia , Surdez/genética , Células Ciliadas Auditivas Internas/metabolismo , Caderinas/metabolismo , Éxons/genética
10.
Hum Mol Genet ; 33(5): 465-474, 2024 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-37988592

RESUMO

Whole genome sequencing (WGS) from large clinically unselected cohorts provides a unique opportunity to assess the penetrance and expressivity of rare and/or known pathogenic mitochondrial variants in population. Using WGS from 179 862 clinically unselected individuals from the UK Biobank, we performed extensive single and rare variant aggregation association analyses of 15 881 mtDNA variants and 73 known pathogenic variants with 15 mitochondrial disease-relevant phenotypes. We identified 12 homoplasmic and one heteroplasmic variant (m.3243A>G) with genome-wide significant associations in our clinically unselected cohort. Heteroplasmic m.3243A>G (MAF = 0.0002, a known pathogenic variant) was associated with diabetes, deafness and heart failure and 12 homoplasmic variants increased aspartate aminotransferase levels including three low-frequency variants (MAF ~0.002 and beta~0.3 SD). Most pathogenic mitochondrial disease variants (n = 66/74) were rare in the population (<1:9000). Aggregated or single variant analysis of pathogenic variants showed low penetrance in unselected settings for the relevant phenotypes, except m.3243A>G. Multi-system disease risk and penetrance of diabetes, deafness and heart failure greatly increased with m.3243A>G level ≥ 10%. The odds ratio of these traits increased from 5.61, 12.3 and 10.1 to 25.1, 55.0 and 39.5, respectively. Diabetes risk with m.3243A>G was further influenced by type 2 diabetes genetic risk. Our study of mitochondrial variation in a large-unselected population identified novel associations and demonstrated that pathogenic mitochondrial variants have lower penetrance in clinically unselected settings. m.3243A>G was an exception at higher heteroplasmy showing a significant impact on health making it a good candidate for incidental reporting.


Assuntos
Surdez , Diabetes Mellitus Tipo 2 , Insuficiência Cardíaca , Doenças Mitocondriais , Humanos , Penetrância , Diabetes Mellitus Tipo 2/genética , DNA Mitocondrial/genética , Doenças Mitocondriais/genética , Surdez/genética , Mutação
11.
PLoS Biol ; 21(6): e3002160, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37368868

RESUMO

Intrinsically generated neural activity propagates through the developing auditory system to promote maturation and refinement of sound processing circuits prior to hearing onset. This early patterned activity is induced by non-sensory supporting cells in the organ of Corti, which are highly interconnected through gap junctions containing connexin 26 (Gjb2). Although loss of function mutations in Gjb2 impair cochlear development and are the most common cause of congenital deafness, it is not known if these variants disrupt spontaneous activity and the developmental trajectory of sound processing circuits in the brain. Here, we show in a new mouse model of Gjb2-mediated congenital deafness that cochlear supporting cells adjacent to inner hair cells (IHCs) unexpectedly retain intercellular coupling and the capacity to generate spontaneous activity, exhibiting only modest deficits prior to hearing onset. Supporting cells lacking Gjb2 elicited coordinated activation of IHCs, leading to coincident bursts of activity in central auditory neurons that will later process similar frequencies of sound. Despite alterations in the structure of the sensory epithelium, hair cells within the cochlea of Gjb2-deficient mice were intact and central auditory neurons could be activated within appropriate tonotopic domains by loud sounds at hearing onset, indicating that early maturation and refinement of auditory circuits was preserved. Only after cessation of spontaneous activity following hearing onset did progressive hair cell degeneration and enhanced auditory neuron excitability manifest. This preservation of cochlear spontaneous neural activity in the absence of connexin 26 may increase the effectiveness of early therapeutic interventions to restore hearing.


Assuntos
Cóclea , Surdez , Camundongos , Animais , Conexina 26 , Cóclea/fisiologia , Células Ciliadas Auditivas/fisiologia , Células Ciliadas Auditivas Internas/fisiologia , Surdez/genética
12.
Proc Natl Acad Sci U S A ; 120(7): e2215423120, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36745780

RESUMO

Due to the ubiquitous nature of language in the environment of infants, how it affects the anatomical structure of the brain language system over the lifespan is not well understood. In this study, we investigated the effects of early language experience on the adult brain by examining anatomical features of individuals born deaf with typical or restricted language experience in early childhood. Twenty-two deaf adults whose primary language was American Sign Language and were first immersed in it at ages ranging from birth to 14 y participated. The control group was 21 hearing non-signers. We acquired T1-weighted magnetic resonance images and used FreeSurfer [B. Fischl, Neuroimage 62, 774-781(2012)] to reconstruct the brain surface. Using an a priori regions of interest (ROI) approach, we identified 17 language and 19 somatomotor ROIs in each hemisphere from the Human Connectome Project parcellation map [M. F. Glasser et al., Nature 536, 171-178 (2016)]. Restricted language experience in early childhood was associated with negative changes in adjusted grey matter volume and/or cortical thickness in bilateral fronto-temporal regions. No evidence of anatomical differences was observed in any of these regions when deaf signers with infant sign language experience were compared with hearing speakers with infant spoken language experience, showing that the effects of early language experience on the brain language system are supramodal.


Assuntos
Surdez , Pré-Escolar , Humanos , Adulto , Surdez/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Idioma , Audição , Língua de Sinais
13.
Proc Natl Acad Sci U S A ; 120(26): e2221744120, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37339214

RESUMO

Functional molecular characterization of the cochlea has mainly been driven by the deciphering of the genetic architecture of sensorineural deafness. As a result, the search for curative treatments, which are sorely lacking in the hearing field, has become a potentially achievable objective, particularly via cochlear gene and cell therapies. To this end, a complete inventory of cochlear cell types, with an in-depth characterization of their gene expression profiles right up to their final differentiation, is indispensable. We therefore generated a single-cell transcriptomic atlas of the mouse cochlea based on an analysis of more than 120,000 cells on postnatal day 8 (P8), during the prehearing period, P12, corresponding to hearing onset, and P20, when cochlear maturation is almost complete. By combining whole-cell and nuclear transcript analyses with extensive in situ RNA hybridization assays, we characterized the transcriptomic signatures covering nearly all cochlear cell types and developed cell type-specific markers. Three cell types were discovered; two of them contribute to the modiolus which houses the primary auditory neurons and blood vessels, and the third one consists in cells lining the scala vestibuli. The results also shed light on the molecular basis of the tonotopic gradient of the biophysical characteristics of the basilar membrane that critically underlies cochlear passive sound frequency analysis. Finally, overlooked expression of deafness genes in several cochlear cell types was also unveiled. This atlas paves the way for the deciphering of the gene regulatory networks controlling cochlear cell differentiation and maturation, essential for the development of effective targeted treatments.


Assuntos
Surdez , Transcriptoma , Animais , Camundongos , Cóclea/fisiologia , Membrana Basilar , Audição/fisiologia , Surdez/metabolismo
14.
PLoS Genet ; 19(1): e1010584, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36656851

RESUMO

Loss or absence of hearing is common at both extremes of human lifespan, in the forms of congenital deafness and age-related hearing loss. While these are often studied separately, there is increasing evidence that their genetic basis is at least partially overlapping. In particular, both common and rare variants in genes associated with monogenic forms of hearing loss also contribute to the more polygenic basis of age-related hearing loss. Here, we directly test this model in the Penn Medicine BioBank-a healthcare system cohort of around 40,000 individuals with linked genetic and electronic health record data. We show that increased burden of predicted deleterious variants in Mendelian hearing loss genes is associated with increased risk and severity of adult-onset hearing loss. As a specific example, we identify one gene-TCOF1, responsible for a syndromic form of congenital hearing loss-in which deleterious variants are also associated with adult-onset hearing loss. We also identify four additional novel candidate genes (COL5A1, HMMR, RAPGEF3, and NNT) in which rare variant burden may be associated with hearing loss. Our results confirm that rare variants in Mendelian hearing loss genes contribute to polygenic risk of hearing loss, and emphasize the utility of healthcare system cohorts to study common complex traits and diseases.


Assuntos
Surdez , Perda Auditiva Neurossensorial , Perda Auditiva , Humanos , Adulto , Surdez/genética , Perda Auditiva/genética , Perda Auditiva Neurossensorial/genética , Herança Multifatorial , Audição , Mutação
15.
PLoS Genet ; 19(11): e1011058, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38011198

RESUMO

Adult-onset progressive hearing loss is a common, complex disease with a strong genetic component. Although to date over 150 genes have been identified as contributing to human hearing loss, many more remain to be discovered, as does most of the underlying genetic diversity. Many different variants have been found to underlie adult-onset hearing loss, but they tend to be rare variants with a high impact upon the gene product. It is likely that combinations of more common, lower impact variants also play a role in the prevalence of the disease. Here we present our exome study of hearing loss in a cohort of 532 older adult volunteers with extensive phenotypic data, including 99 older adults with normal hearing, an important control set. Firstly, we carried out an outlier analysis to identify genes with a high variant load in older adults with hearing loss compared to those with normal hearing. Secondly, we used audiometric threshold data to identify individual variants which appear to contribute to different threshold values. We followed up these analyses in a second cohort. Using these approaches, we identified genes and variants linked to better hearing as well as those linked to worse hearing. These analyses identified some known deafness genes, demonstrating proof of principle of our approach. However, most of the candidate genes are novel associations with hearing loss. While the results support the suggestion that genes responsible for severe deafness may also be involved in milder hearing loss, they also suggest that there are many more genes involved in hearing which remain to be identified. Our candidate gene lists may provide useful starting points for improved diagnosis and drug development.


Assuntos
Surdez , Perda Auditiva Neurossensorial , Perda Auditiva , Humanos , Idoso , Perda Auditiva Neurossensorial/genética , Sequenciamento do Exoma , Perda Auditiva/genética , Audição , Surdez/genética , Linhagem , Mutação
16.
J Neurosci ; 44(28)2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38830759

RESUMO

Congenital single-sided deafness (SSD) leads to an aural preference syndrome that is characterized by overrepresentation of the hearing ear in the auditory system. Cochlear implantation (CI) of the deaf ear is an effective treatment for SSD. However, the newly introduced auditory input in congenital SSD often does not reach expectations in late-implanted CI recipients with respect to binaural hearing and speech perception. In a previous study, a reduction of the interaural time difference (ITD) sensitivity has been shown in unilaterally congenitally deaf cats (uCDCs). In the present study, we focused on the interaural level difference (ILD) processing in the primary auditory cortex. The uCDC group was compared with hearing cats (HCs) and bilaterally congenitally deaf cats (CDCs). The ILD representation was reorganized, replacing the preference for the contralateral ear with a preference for the hearing ear, regardless of the cortical hemisphere. In accordance with the previous study, uCDCs were less sensitive to interaural time differences than HCs, resulting in unmodulated ITD responses, thus lacking directional information. Such incongruent ITDs and ILDs cannot be integrated for binaural sound source localization. In normal hearing, the predominant effect of each ear is excitation of the auditory cortex in the contralateral cortical hemisphere and inhibition in the ipsilateral hemisphere. In SSD, however, auditory pathways reorganized such that the hearing ear produced greater excitation in both cortical hemispheres and the deaf ear produced weaker excitation and preserved inhibition in both cortical hemispheres.


Assuntos
Córtex Auditivo , Implante Coclear , Sinais (Psicologia) , Perda Auditiva Unilateral , Localização de Som , Gatos , Animais , Localização de Som/fisiologia , Perda Auditiva Unilateral/fisiopatologia , Implante Coclear/métodos , Córtex Auditivo/fisiopatologia , Feminino , Masculino , Estimulação Acústica/métodos , Lateralidade Funcional/fisiologia , Surdez/fisiopatologia , Surdez/congênito , Surdez/cirurgia
17.
J Neurosci ; 44(16)2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38395618

RESUMO

Pure-tone audiograms often poorly predict elderly humans' ability to communicate in everyday complex acoustic scenes. Binaural processing is crucial for discriminating sound sources in such complex acoustic scenes. The compromised perception of communication signals presented above hearing threshold has been linked to both peripheral and central age-related changes in the auditory system. Investigating young and old Mongolian gerbils of both sexes, an established model for human hearing, we demonstrate age-related supra-threshold deficits in binaural hearing using behavioral, electrophysiological, anatomical, and imaging methods. Binaural processing ability was measured as the binaural masking level difference (BMLD), an established measure in human psychophysics. We tested gerbils behaviorally with "virtual headphones," recorded single-unit responses in the auditory midbrain and evaluated gross midbrain and cortical responses using positron emission tomography (PET) imaging. Furthermore, we obtained additional measures of auditory function based on auditory brainstem responses, auditory-nerve synapse counts, and evidence for central inhibitory processing revealed by PET. BMLD deteriorates already in middle-aged animals having normal audiometric thresholds and is even worse in old animals with hearing loss. The magnitude of auditory brainstem response measures related to auditory-nerve function and binaural processing in the auditory brainstem also deteriorate. Furthermore, central GABAergic inhibition is affected by age. Because the number of synapses in the apical turn of the inner ear was not reduced in middle-aged animals, we conclude that peripheral synaptopathy contributes little to binaural processing deficits. Exploratory analyses suggest increased hearing thresholds, altered binaural processing in the brainstem and changed central GABAergic inhibition as potential contributors.


Assuntos
Surdez , Perda Auditiva , Masculino , Idoso , Pessoa de Meia-Idade , Feminino , Animais , Humanos , Gerbillinae , Audição/fisiologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Limiar Auditivo , Percepção Auditiva/fisiologia , Estimulação Acústica
18.
J Neurosci ; 44(13)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38383498

RESUMO

Within the intricate matrices of cognitive neuroscience, auditory deprivation acts as a catalyst, propelling a cascade of neuroanatomical adjustments that have, until now, been suboptimally articulated in extant literature. Addressing this gap, our study harnesses high-resolution 3 T MRI modalities to unveil the multifaceted cortical transformations that emerge in tandem with congenital auditory deficits. We conducted a rigorous cortical surface analysis on a cohort of 90 congenitally deaf individuals, systematically compared with 90 normoacoustic controls. Our sample encompassed both male and female participants, ensuring a gender-inclusive perspective in our analysis. Expected alterations within prototypical auditory domains were evident, but our findings transcended these regions, spotlighting modifications dispersed across a gamut of cortical and subcortical structures, thereby epitomizing the cerebral adaptive dynamics to sensory voids. Crucially, the study's innovative methodology integrated two pivotal variables: the duration of auditory deprivation and the extent of sign language immersion. By intersecting these metrics with structural changes, our analysis unveiled nuanced layers of cortical reconfigurations, elucidating a more granulated understanding of neural plasticity. This intersectional approach bestows a unique advantage, allowing for a discerning exploration into how varying durations of sensory experience and alternative communication modalities modulate the brain's morphological terrain. In encapsulating the synergy of neuroimaging finesse and incisive scientific rigor, this research not only broadens the current understanding of adaptive neural mechanisms but also paves the way for tailored therapeutic strategies, finely attuned to individual auditory histories and communicative repertoires.


Assuntos
Córtex Auditivo , Surdez , Humanos , Masculino , Feminino , Imageamento por Ressonância Magnética , Córtex Auditivo/diagnóstico por imagem , Plasticidade Neuronal
19.
J Biol Chem ; 300(5): 107235, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38552739

RESUMO

Defects in mitochondrial RNA metabolism have been linked to sensorineural deafness that often occurs as a consequence of damaged or deficient inner ear hair cells. In this report, we investigated the molecular mechanism underlying a deafness-associated tRNAPhe 593T > C mutation that changed a highly conserved uracil to cytosine at position 17 of the DHU-loop. The m.593T > C mutation altered tRNAPhe structure and function, including increased melting temperature, resistance to S1 nuclease-mediated digestion, and conformational changes. The aberrant tRNA metabolism impaired mitochondrial translation, which was especially pronounced by decreases in levels of ND1, ND5, CYTB, CO1, and CO3 harboring higher numbers of phenylalanine. These alterations resulted in aberrant assembly, instability, and reduced activities of respiratory chain enzyme complexes I, III, IV, and intact supercomplexes overall. Furthermore, we found that the m.593T > C mutation caused markedly diminished membrane potential, and increased the production of reactive oxygen species in the mutant cell lines carrying the m.593T > C mutation. These mitochondrial dysfunctions led to the mitochondrial dynamic imbalance via increasing fission with abnormal mitochondrial morphology. Excessive fission impaired the process of autophagy including the initiation phase, formation, and maturation of the autophagosome. In particular, the m.593T > C mutation upregulated the PARKIN-dependent mitophagy pathway. These alterations promoted an intrinsic apoptotic process for the removal of damaged cells. Our findings provide critical insights into the pathophysiology of maternally inherited deafness arising from tRNA mutation-induced defects in mitochondrial and cellular integrity.


Assuntos
Surdez , Mitocôndrias , RNA de Transferência de Fenilalanina , Humanos , Autofagia , Surdez/genética , Surdez/metabolismo , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Mitocôndrias/genética , Mitocôndrias/patologia , Dinâmica Mitocondrial , Mutação , Espécies Reativas de Oxigênio/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , RNA de Transferência de Fenilalanina/genética
20.
J Biol Chem ; 300(7): 107474, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38879011

RESUMO

Hearing, the ability to sense sounds, and the processing of auditory information are important for perception of the world. Mice lacking expression of neuroplastin (Np), a type-1 transmembrane glycoprotein, display deafness, multiple cognitive deficiencies, and reduced expression of plasma membrane calcium (Ca2+) ATPases (PMCAs) in cochlear hair cells and brain neurons. In this study, we transferred the deafness causing missense mutations pitch (C315S) and audio-1 (I122N) into human Np (hNp) constructs and investigated their effects at the molecular and cellular levels. Computational molecular dynamics show that loss of the disulfide bridge in hNppitch causes structural destabilization of immunoglobulin-like domain (Ig) III and that the novel asparagine in hNpaudio-1 results in steric constraints and an additional N-glycosylation site in IgII. Additional N-glycosylation of hNpaudio-1 was confirmed by PNGaseF treatment. In comparison to hNpWT, transfection of hNppitch and hNpaudio-1 into HEK293T cells resulted in normal mRNA levels but reduced the Np protein levels and their cell surface expression due to proteasomal/lysosomal degradation. Furthermore, hNppitch and hNpaudio-1 failed to promote exogenous PMCA levels in HEK293T cells. In hippocampal neurons, expression of additional hNppitch or hNpaudio-1 was less efficient than hNpWT to elevate endogenous PMCA levels and to accelerate the restoration of basal Ca2+ levels after electrically evoked Ca2+ transients. We propose that mutations leading to pathological Np variants, as exemplified here by the deafness causing Np mutants, can affect Np-dependent Ca2+ regulatory mechanisms and may potentially cause intellectual and cognitive deficits in humans.


Assuntos
Encéfalo , Cálcio , Surdez , Glicoproteínas de Membrana , Mutação de Sentido Incorreto , Neurônios , ATPases Transportadoras de Cálcio da Membrana Plasmática , Humanos , Surdez/metabolismo , Surdez/genética , Surdez/patologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Neurônios/metabolismo , Células HEK293 , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Cálcio/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Membrana Celular/metabolismo , Camundongos , Glicosilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA