Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 111(3): 614-617, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38330941

RESUMEN

Age-related hearing loss (ARHL) is a major health concern among the elderly population. It is hoped that increasing our understanding of its underlying pathophysiological processes will lead to the development of novel therapies. Recent genome-wide association studies (GWASs) discovered a few dozen genetic variants in association with elevated risk for ARHL. Integrated analysis of GWAS results and transcriptomics data is a powerful approach for elucidating specific cell types that are involved in disease pathogenesis. Intriguingly, recent studies that applied such bioinformatics approaches to ARHL resulted in disagreeing findings as for the key cell types that are most strongly linked to the genetic pathogenesis of ARHL. These conflicting studies pointed either to cochlear sensory epithelial or to stria vascularis cells as the cell types most prominently involved in the genetic basis of ARHL. Seeking to resolve this discrepancy, we integrated the analysis of four ARHL GWAS datasets with four independent inner-ear single-cell RNA-sequencing datasets. Our analysis clearly points to the cochlear sensory epithelial cells as the key cells for the genetic predisposition to ARHL. We also explain the limitation of the bioinformatics analysis performed by previous studies that led to missing the enrichment for ARHL GWAS signal in sensory epithelial cells. Collectively, we show that cochlear epithelial cells, not stria vascularis cells, are the main inner-ear cells related to the genetic pathogenesis of ARHL.


Asunto(s)
Presbiacusia , Estría Vascular , Anciano , Humanos , Estría Vascular/patología , Estudio de Asociación del Genoma Completo , Cóclea/patología , Presbiacusia/genética , Presbiacusia/patología , Epitelio/patología
2.
Nature ; 598(7879): 103-110, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616066

RESUMEN

Single-cell transcriptomics can provide quantitative molecular signatures for large, unbiased samples of the diverse cell types in the brain1-3. With the proliferation of multi-omics datasets, a major challenge is to validate and integrate results into a biological understanding of cell-type organization. Here we generated transcriptomes and epigenomes from more than 500,000 individual cells in the mouse primary motor cortex, a structure that has an evolutionarily conserved role in locomotion. We developed computational and statistical methods to integrate multimodal data and quantitatively validate cell-type reproducibility. The resulting reference atlas-containing over 56 neuronal cell types that are highly replicable across analysis methods, sequencing technologies and modalities-is a comprehensive molecular and genomic account of the diverse neuronal and non-neuronal cell types in the mouse primary motor cortex. The atlas includes a population of excitatory neurons that resemble pyramidal cells in layer 4 in other cortical regions4. We further discovered thousands of concordant marker genes and gene regulatory elements for these cell types. Our results highlight the complex molecular regulation of cell types in the brain and will directly enable the design of reagents to target specific cell types in the mouse primary motor cortex for functional analysis.


Asunto(s)
Epigenómica , Perfilación de la Expresión Génica , Corteza Motora/citología , Neuronas/clasificación , Análisis de la Célula Individual , Transcriptoma , Animales , Atlas como Asunto , Conjuntos de Datos como Asunto , Epigénesis Genética , Femenino , Masculino , Ratones , Corteza Motora/anatomía & histología , Neuronas/citología , Neuronas/metabolismo , Especificidad de Órganos , Reproducibilidad de los Resultados
3.
PLoS Biol ; 21(6): e3002133, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37390046

RESUMEN

Characterizing cellular diversity at different levels of biological organization and across data modalities is a prerequisite to understanding the function of cell types in the brain. Classification of neurons is also essential to manipulate cell types in controlled ways and to understand their variation and vulnerability in brain disorders. The BRAIN Initiative Cell Census Network (BICCN) is an integrated network of data-generating centers, data archives, and data standards developers, with the goal of systematic multimodal brain cell type profiling and characterization. Emphasis of the BICCN is on the whole mouse brain with demonstration of prototype feasibility for human and nonhuman primate (NHP) brains. Here, we provide a guide to the cellular and spatial approaches employed by the BICCN, and to accessing and using these data and extensive resources, including the BRAIN Cell Data Center (BCDC), which serves to manage and integrate data across the ecosystem. We illustrate the power of the BICCN data ecosystem through vignettes highlighting several BICCN analysis and visualization tools. Finally, we present emerging standards that have been developed or adopted toward Findable, Accessible, Interoperable, and Reusable (FAIR) neuroscience. The combined BICCN ecosystem provides a comprehensive resource for the exploration and analysis of cell types in the brain.


Asunto(s)
Encéfalo , Neurociencias , Animales , Humanos , Ratones , Ecosistema , Neuronas
4.
Annu Rev Genomics Hum Genet ; 23: 275-299, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35667089

RESUMEN

Current estimates suggest that nearly half a billion people worldwide are affected by hearing loss. Because of the major psychological, social, economic, and health ramifications, considerable efforts have been invested in identifying the genes and molecular pathways involved in hearing loss, whether genetic or environmental, to promote prevention, improve rehabilitation, and develop therapeutics. Genomic sequencing technologies have led to the discovery of genes associated with hearing loss. Studies of the transcriptome and epigenome of the inner ear have characterized key regulators and pathways involved in the development of the inner ear and have paved the way for their use in regenerative medicine. In parallel, the immense preclinical success of using viral vectors for gene delivery in animal models of hearing loss has motivated the industry to work on translating such approaches into the clinic. Here, we review the recent advances in the genomics of auditory function and dysfunction, from patient diagnostics to epigenetics and gene therapy.


Asunto(s)
Sordera , Oído Interno , Pérdida Auditiva , Animales , Sordera/metabolismo , Sordera/terapia , Oído Interno/metabolismo , Terapia Genética , Genómica , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Humanos
5.
Am J Hum Genet ; 109(6): 1077-1091, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35580588

RESUMEN

Hearing loss is one of the top contributors to years lived with disability and is a risk factor for dementia. Molecular evidence on the cellular origins of hearing loss in humans is growing. Here, we performed a genome-wide association meta-analysis of clinically diagnosed and self-reported hearing impairment on 723,266 individuals and identified 48 significant loci, 10 of which are novel. A large proportion of associations comprised missense variants, half of which lie within known familial hearing loss loci. We used single-cell RNA-sequencing data from mouse cochlea and brain and mapped common-variant genomic results to spindle, root, and basal cells from the stria vascularis, a structure in the cochlea necessary for normal hearing. Our findings indicate the importance of the stria vascularis in the mechanism of hearing impairment, providing future paths for developing targets for therapeutic intervention in hearing loss.


Asunto(s)
Sordera , Pérdida Auditiva , Animales , Cóclea , Estudio de Asociación del Genoma Completo , Pérdida Auditiva/genética , Humanos , Ratones , Estría Vascular
6.
Nucleic Acids Res ; 51(D1): D1075-D1085, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36318260

RESUMEN

Scalable technologies to sequence the transcriptomes and epigenomes of single cells are transforming our understanding of cell types and cell states. The Brain Research through Advancing Innovative Neurotechnologies (BRAIN) Initiative Cell Census Network (BICCN) is applying these technologies at unprecedented scale to map the cell types in the mammalian brain. In an effort to increase data FAIRness (Findable, Accessible, Interoperable, Reusable), the NIH has established repositories to make data generated by the BICCN and related BRAIN Initiative projects accessible to the broader research community. Here, we describe the Neuroscience Multi-Omic Archive (NeMO Archive; nemoarchive.org), which serves as the primary repository for genomics data from the BRAIN Initiative. Working closely with other BRAIN Initiative researchers, we have organized these data into a continually expanding, curated repository, which contains transcriptomic and epigenomic data from over 50 million brain cells, including single-cell genomic data from all of the major regions of the adult and prenatal human and mouse brains, as well as substantial single-cell genomic data from non-human primates. We make available several tools for accessing these data, including a searchable web portal, a cloud-computing interface for large-scale data processing (implemented on Terra, terra.bio), and a visualization and analysis platform, NeMO Analytics (nemoanalytics.org).


Asunto(s)
Encéfalo , Bases de Datos Genéticas , Epigenómica , Multiómica , Transcriptoma , Animales , Ratones , Genómica , Mamíferos , Primates , Encéfalo/citología , Encéfalo/metabolismo
7.
PLoS Biol ; 19(11): e3001445, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34758021

RESUMEN

Cochlear supporting cells (SCs) are glia-like cells critical for hearing function. In the neonatal cochlea, the greater epithelial ridge (GER) is a mitotically quiescent and transient organ, which has been shown to nonmitotically regenerate SCs. Here, we ablated Lgr5+ SCs using Lgr5-DTR mice and found mitotic regeneration of SCs by GER cells in vivo. With lineage tracing, we show that the GER houses progenitor cells that robustly divide and migrate into the organ of Corti to replenish ablated SCs. Regenerated SCs display coordinated calcium transients, markers of the SC subtype inner phalangeal cells, and survive in the mature cochlea. Via RiboTag, RNA-sequencing, and gene clustering algorithms, we reveal 11 distinct gene clusters comprising markers of the quiescent and damaged GER, and damage-responsive genes driving cell migration and mitotic regeneration. Together, our study characterizes GER cells as mitotic progenitors with regenerative potential and unveils their quiescent and damaged translatomes.


Asunto(s)
Linaje de la Célula/genética , Cóclea/citología , Estudios de Asociación Genética , Mitosis , Biosíntesis de Proteínas , Regeneración/genética , Células Madre/citología , Células Madre/metabolismo , Animales , Diferenciación Celular , Supervivencia Celular , Células Epiteliales/citología , Regulación de la Expresión Génica , Integrasas/metabolismo , Ratones , Familia de Multigenes , Receptores Acoplados a Proteínas G/metabolismo
8.
Nature ; 563(7733): 696-700, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30464345

RESUMEN

The sensory cells that are responsible for hearing include the cochlear inner hair cells (IHCs) and outer hair cells (OHCs), with the OHCs being necessary for sound sensitivity and tuning1. Both cell types are thought to arise from common progenitors; however, our understanding of the factors that control the fate of IHCs and OHCs remains limited. Here we identify Ikzf2 (which encodes Helios) as an essential transcription factor in mice that is required for OHC functional maturation and hearing. Helios is expressed in postnatal mouse OHCs, and in the cello mouse model a point mutation in Ikzf2 causes early-onset sensorineural hearing loss. Ikzf2cello/cello OHCs have greatly reduced prestin-dependent electromotile activity, a hallmark of OHC functional maturation, and show reduced levels of crucial OHC-expressed genes such as Slc26a5 (which encodes prestin) and Ocm. Moreover, we show that ectopic expression of Ikzf2 in IHCs: induces the expression of OHC-specific genes; reduces the expression of canonical IHC genes; and confers electromotility to IHCs, demonstrating that Ikzf2 can partially shift the IHC transcriptome towards an OHC-like identity.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Células Ciliadas Auditivas Externas/citología , Células Ciliadas Auditivas Externas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Transcriptoma/genética , Animales , Secuencia de Bases , Biomarcadores/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
9.
Development ; 147(17)2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917668

RESUMEN

Despite the known importance of the transcription factors ATOH1, POU4F3 and GFI1 in hair cell development and regeneration, their downstream transcriptional cascades in the inner ear remain largely unknown. Here, we have used Gfi1cre;RiboTag mice to evaluate changes to the hair cell translatome in the absence of GFI1. We identify a systematic downregulation of hair cell differentiation genes, concomitant with robust upregulation of neuronal genes in the GFI1-deficient hair cells. This includes increased expression of neuronal-associated transcription factors (e.g. Pou4f1) as well as transcription factors that serve dual roles in hair cell and neuronal development (e.g. Neurod1, Atoh1 and Insm1). We further show that the upregulated genes are consistent with the NEUROD1 regulon and are normally expressed in hair cells prior to GFI1 onset. Additionally, minimal overlap of differentially expressed genes in auditory and vestibular hair cells suggests that GFI1 serves different roles in these systems. From these data, we propose a dual mechanism for GFI1 in promoting hair cell development, consisting of repression of neuronal-associated genes as well as activation of hair cell-specific genes required for normal functional maturation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Células Ciliadas Auditivas Internas/metabolismo , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al ADN/genética , Células Ciliadas Auditivas Internas/citología , Ratones , Ratones Transgénicos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción Brn-3A/genética , Factor de Transcripción Brn-3A/metabolismo , Factores de Transcripción/genética
10.
Ear Hear ; 44(1): 10-27, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36384870

RESUMEN

There is robust evidence that sex (biological) and gender (behavioral/social) differences influence hearing loss risk and outcomes. These differences are noted for animals and humans-in the occurrence of hearing loss, hearing loss progression, and response to interventions. Nevertheless, many studies have not reported or disaggregated data by sex or gender. This article describes the influence of sex-linked biology (specifically sex-linked hormones) and gender on hearing and hearing interventions, including the role of sex-linked biology and gender in modifying the association between risk factors and hearing loss, and the effects of hearing loss on quality of life and functioning. Most prevalence studies indicate that hearing loss begins earlier and is more common and severe among men than women. Intrinsic sex-linked biological differences in the auditory system may account, in part, for the predominance of hearing loss in males. Sex- and gender-related differences in the effects of noise exposure or cardiovascular disease on the auditory system may help explain some of these differences in the prevalence of hearing loss. Further still, differences in hearing aid use and uptake, and the effects of hearing loss on health may also vary by sex and gender. Recognizing that sex-linked biology and gender are key determinants of hearing health, the present review concludes by emphasizing the importance of a well-developed research platform that proactively measures and assesses sex- and gender-related differences in hearing, including in understudied populations. Such research focus is necessary to advance the field of hearing science and benefit all members of society.


Asunto(s)
Sordera , Pérdida Auditiva , Masculino , Humanos , Femenino , Calidad de Vida , Pérdida Auditiva/epidemiología , Pérdida Auditiva/rehabilitación , Pruebas Auditivas , Audición , Biología
11.
PLoS Genet ; 16(9): e1009025, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32986727

RESUMEN

Age-related hearing impairment (ARHI), one of the most common medical conditions, is strongly heritable, yet its genetic causes remain largely unknown. We conducted a meta-analysis of GWAS summary statistics from multiple hearing-related traits in the UK Biobank (n = up to 330,759) and identified 31 genome-wide significant risk loci for self-reported hearing difficulty (p < 5x10-8), of which eight have not been reported previously in the peer-reviewed literature. We investigated the regulatory and cell specific expression for these loci by generating mRNA-seq, ATAC-seq, and single-cell RNA-seq from cells in the mouse cochlea. Risk-associated genes were most strongly enriched for expression in cochlear epithelial cells, as well as for genes related to sensory perception and known Mendelian deafness genes, supporting their relevance to auditory function. Regions of the human genome homologous to open chromatin in epithelial cells from the mouse were strongly enriched for heritable risk for hearing difficulty, even after adjusting for baseline effects of evolutionary conservation and cell-type non-specific regulatory regions. Epigenomic and statistical fine-mapping most strongly supported 50 putative risk genes. Of these, 39 were expressed robustly in mouse cochlea and 16 were enriched specifically in sensory hair cells. These results reveal new risk loci and risk genes for hearing difficulty and suggest an important role for altered gene regulation in the cochlear sensory epithelium.


Asunto(s)
Cóclea/citología , Sitios Genéticos , Predisposición Genética a la Enfermedad , Pérdida Auditiva/genética , Adulto , Animales , Bancos de Muestras Biológicas , Cromatina/genética , Estudios de Cohortes , Epigenoma , Células Epiteliales/fisiología , Femenino , Estudio de Asociación del Genoma Completo , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/fisiología , Humanos , Ratones Endogámicos ICR , Ratones Endogámicos , Polimorfismo de Nucleótido Simple , Análisis de la Célula Individual , Reino Unido
12.
Hum Genet ; 141(3-4): 319-322, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35235019

RESUMEN

High-throughput cell type-specific multi-omic analyses have advanced our understanding of inner ear biology in an unprecedented way. The full benefit of these data, however, is reached from their re-use. Successful re-use of data requires identifying the natural users and ensuring proper data democratization and federation for their seamless and meaningful access. Here we discuss universal challenges in access and re-use of multi-omic data, possible solutions, and introduce the gEAR (the gene Expression Analysis Resource, umgear.org)-a tool for multi-omic data visualization, sharing and access for the ear field.


Asunto(s)
Genómica , Audición , Humanos
13.
Mol Psychiatry ; 26(6): 1846-1859, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32366954

RESUMEN

Motor stereotypies occurring in early-onset neuropsychiatric diseases are associated with dysregulated basal ganglia direct-pathway activity. Disruptions in network connectivity through impaired neuronal structure have been implicated in both rodents and humans. However, the neurobiological mechanisms leading to direct-pathway neuron disconnectivity in stereotypy remain poorly understood. We have a mouse line with Tropomyosin receptor kinase B (TrkB) receptor deletion from D1-expressing cells (D1-Cre-flTrkB) in which a subset of animals shows repetitive rotations and head tics with juvenile onset. Here we demonstrate these behaviors may be associated with abnormal direct-pathway activity by reducing rotations using chemogenetic inhibition of dorsal striatum D1-medium spiny neurons (D1-MSNs) in both juvenile and young-adult mice. Taking advantage of phenotypical differences in animals with similar genotypes, we then interrogated the D1-MSN specific translatome associated with repetitive behavior by using RNA sequencing of ribosome-associated mRNA. Detailed translatome analysis followed by multiplexed gene expression assessment revealed profound alterations in neuronal projection and synaptic structure related genes in stereotypy mice. Examination of neuronal morphology demonstrated dendritic atrophy and dendritic spine loss in dorsal striatum D1-MSNs from mice with repetitive behavior. Together, our results uncover phenotype-specific molecular alterations in D1-MSNs that relate to morphological adaptations in mice displaying stereotypy behavior.


Asunto(s)
Receptores de Dopamina D1 , Receptores de Dopamina D2 , Animales , Cuerpo Estriado/metabolismo , Individualidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo
14.
Hum Mol Genet ; 28(13): 2212-2223, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31220269

RESUMEN

Alström syndrome (OMIM #203800) is an autosomal recessive obesity ciliopathy caused by loss-of-function mutations in the ALMS1 gene. In addition to multi-organ dysfunction, such as cardiomyopathy, retinal degeneration and renal dysfunction, the disorder is characterized by high rates of obesity, insulin resistance and early-onset type 2 diabetes mellitus (T2DM). To investigate the underlying mechanisms of T2DM phenotypes, we generated a loss-of-function deletion of alms1 in the zebrafish. We demonstrate conservation of hallmark clinical characteristics alongside metabolic syndrome phenotypes, including a propensity for obesity and fatty livers, hyperinsulinemia and glucose response defects. Gene expression changes in ß-cells isolated from alms1-/- mutants revealed changes consistent with insulin hypersecretion and glucose sensing failure, which were corroborated in cultured murine ß-cells lacking Alms1. We also found evidence of defects in peripheral glucose uptake and concomitant hyperinsulinemia in the alms1-/- animals. We propose a model in which hyperinsulinemia is the primary and causative defect underlying generation of T2DM associated with alms1 deficiency. These observations support the alms1 loss-of-function zebrafish mutant as a monogenic model for mechanistic interrogation of T2DM phenotypes.


Asunto(s)
Síndrome de Alstrom/genética , Diabetes Mellitus Tipo 2/genética , Resistencia a la Insulina/genética , Insuficiencia Renal/genética , Degeneración Retiniana/genética , Pez Cebra/genética , Síndrome de Alstrom/fisiopatología , Animales , Animales Modificados Genéticamente , Línea Celular , Modelos Animales de Enfermedad , Intolerancia a la Glucosa , Hiperinsulinismo/genética , Células Secretoras de Insulina/metabolismo , Ratones , Modelos Biológicos , Obesidad/genética , Fenotipo , Pez Cebra/embriología
15.
Int J Mol Sci ; 22(22)2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34830090

RESUMEN

Recent studies have identified sex-differences in auditory physiology and in the susceptibility to noise-induced hearing loss (NIHL). We hypothesize that 17ß-estradiol (E2), a known modulator of auditory physiology, may underpin sex-differences in the response to noise trauma. Here, we gonadectomized B6CBAF1/J mice and used a combination of electrophysiological and histological techniques to study the effects of estrogen replacement on peripheral auditory physiology in the absence of noise exposure and on protection from NIHL. Functional analysis of auditory physiology in gonadectomized female mice revealed that E2-treatment modulated the peripheral response to sound in the absence of changes to the endocochlear potential compared to vehicle-treatment. E2-replacement in gonadectomized female mice protected against hearing loss following permanent threshold shift (PTS)- and temporary threshold shift (TTS)-inducing noise exposures. Histological analysis of the cochlear tissue revealed that E2-replacement mitigated outer hair cell loss and cochlear synaptopathy following noise exposure compared to vehicle-treatment. Lastly, using fluorescent in situ hybridization, we demonstrate co-localization of estrogen receptor-2 with type-1C, high threshold spiral ganglion neurons, suggesting that the observed protection from cochlear synaptopathy may occur through E2-mediated preservation of these neurons. Taken together, these data indicate the estrogen signaling pathways may be harnessed for the prevention and treatment of NIHL.


Asunto(s)
Cóclea , Estradiol/farmacología , Potenciales Evocados Auditivos/efectos de los fármacos , Pérdida Auditiva Provocada por Ruido , Animales , Cóclea/metabolismo , Cóclea/patología , Cóclea/fisiopatología , Femenino , Pérdida Auditiva Provocada por Ruido/metabolismo , Pérdida Auditiva Provocada por Ruido/patología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Pérdida Auditiva Provocada por Ruido/prevención & control , Ratones , Ovariectomía
16.
J Neurosci ; 39(33): 6425-6438, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31209173

RESUMEN

During nervous system development, axons often undergo elaborate changes in branching patterns before circuits have achieved their mature patterns of innervation. In the auditory system, type I spiral ganglion neurons (SGNs) project their peripheral axons into the cochlear epithelium and then undergo a process of branch refinement before forming synapses with sensory hair cells. Here, we report that Semaphorin-5B (Sema5B) acts as an important mediator of this process. During cochlear development in mouse, immature hair cells express Sema5B, whereas the SGNs express both PlexinA1 and PlexinA3, which are known Sema5B receptors. In these studies, genetic sparse labeling and three-dimensional reconstruction techniques were leveraged to determine the morphologies of individual type I SGNs after manipulations of Sema5B signaling. Treating cultured mouse cochleae with Sema5B-Fc (to activate Plexin-As) led to type I SGNs with less numerous, but longer terminal branches. Conversely, cochleae from Sema5b knock-out mice showed type I SGNs with more numerous, but shorter terminal branches. In addition, conditional loss of Plxna1 in SGNs (using Bhlhb5Cre) led to increased type I SGN branching, suggesting that PlexinA1 normally responds to Sema5B in this process. In these studies, mice of either sex were used. The data presented here suggest that Sema5B-PlexinA1 signaling limits SGN terminal branch numbers without causing axonal repulsion, which is a role that distinguishes Sema5B from other Semaphorins in cochlear development.SIGNIFICANCE STATEMENT The sensorineural components of the cochlea include hair cells, which respond mechanically to sound waves, and afferent spiral ganglion neurons (SGNs), which respond to glutamate released by hair cells and transmit auditory information into the CNS. An important component of synapse formation in the cochlea is a process of SGN "debranching" whereby SGNs lose extraneous branches before developing unramified bouton endings that contact the hair cells. In this work, we have found that the transmembrane ligand Semaphorin-5B and its receptor PlexinA1 regulate the debranching process. The results in this report provide new knowledge regarding the molecular control of cochlear afferent innervation.


Asunto(s)
Neurogénesis/fisiología , Neuronas/metabolismo , Semaforinas/metabolismo , Ganglio Espiral de la Cóclea/embriología , Animales , Femenino , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Ganglio Espiral de la Cóclea/metabolismo
17.
Clin Genet ; 98(4): 353-364, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33111345

RESUMEN

Mutations in more than 150 genes are responsible for inherited hearing loss, with thousands of different, severe causal alleles that vary among populations. The Israeli Jewish population includes communities of diverse geographic origins, revealing a wide range of deafness-associated variants and enabling clinical characterization of the associated phenotypes. Our goal was to identify the genetic causes of inherited hearing loss in this population, and to determine relationships among genotype, phenotype, and ethnicity. Genomic DNA samples from informative relatives of 88 multiplex families, all of self-identified Jewish ancestry, with either non-syndromic or syndromic hearing loss, were sequenced for known and candidate deafness genes using the HEar-Seq gene panel. The genetic causes of hearing loss were identified for 60% of the families. One gene was encountered for the first time in human hearing loss: ATOH1 (Atonal), a basic helix-loop-helix transcription factor responsible for autosomal dominant progressive hearing loss in a five-generation family. Our results show that genomic sequencing with a gene panel dedicated to hearing loss is effective for genetic diagnoses in a diverse population. Comprehensive sequencing enables well-informed genetic counseling and clinical management by medical geneticists, otolaryngologists, audiologists, and speech therapists and can be integrated into newborn screening for deafness.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sordera/genética , Predisposición Genética a la Enfermedad , Pérdida Auditiva/genética , Adolescente , Adulto , Niño , Preescolar , Sordera/epidemiología , Sordera/patología , Femenino , Estudios de Asociación Genética , Pérdida Auditiva/epidemiología , Pérdida Auditiva/patología , Humanos , Israel/epidemiología , Judíos/genética , Masculino , Linaje , Adulto Joven
18.
Ear Hear ; 41(3): 640-651, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31702596

RESUMEN

OBJECTIVES: Cochlear implants (CIs) are considered a safe and effective intervention for more severe degrees of hearing loss in adults of all ages. Although older CI users ≥65 years of age can obtain large benefits in speech understanding from a CI, there is a growing body of literature suggesting that older CI users may not perform as well as younger CI users. One reason for this potential age-related limitation could be that default CI stimulation settings are not optimal for older CI users. The goal of this study was to determine whether improvements in speech understanding were possible when CI users were programmed with nondefault stimulation rates and to determine whether lower-than-default stimulation rates improved older CI users' speech understanding. DESIGN: Sentence recognition was measured acutely using different stimulation rates in 37 CI users ranging in age from 22 to 87 years. Maps were created using rates of 500, 720, 900, and 1200 pulses per second (pps) for each subject. An additional map using a rate higher than 1200 pps was also created for individuals who used a higher rate in their clinical processors. Thus, the clinical rate of each subject was also tested, including non-default rates above 1200 pps for Cochlear users and higher rates consistent with the manufacturer defaults for subjects implanted with Advanced Bionics and Med-El devices. Speech understanding performance was evaluated at each stimulation rate using AzBio and Perceptually Robust English Sentence Test Open-set (PRESTO) sentence materials tested in quiet and in noise. RESULTS: For Cochlear-brand users, speech understanding performance using non-default rates was slightly poorer when compared with the default rate (900 pps). However, this effect was offset somewhat by age, in which older subjects were able to maintain comparable performance using a 500-pps map compared with the default rate map when listening to the more difficult PRESTO sentence material. Advanced Bionics and Med-El users showed modest improvements in their overall performance using 720 pps compared with the default rate (>1200 pps). On the individual-subject level, 10 subjects (11 ears) showed a significant effect of stimulation rate, with 8 of those ears performing best with a lower-than-default rate. CONCLUSIONS: Results suggest that default stimulation rates are likely sufficient for many CI users, but some CI users at any age can benefit from a lower-than-default rate. Future work that provides experience with novel rates in everyday life has the potential to identify more individuals whose performance could be improved with changes to stimulation rate.


Asunto(s)
Implantación Coclear , Implantes Cocleares , Sordera , Percepción del Habla , Adulto , Anciano , Humanos , Habla
20.
J Acoust Soc Am ; 146(5): 4007, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31795683

RESUMEN

Acoustic trauma is a feature of the industrial age, in general, and mechanized warfare, in particular. Noise-induced hearing loss (NIHL) and tinnitus have been the number 1 and number 2 disabilities at U.S. Veterans hospitals since 2006. In a reversal of original protocols to identify candidate genes associated with monogenic deafness disorders, unbiased genome-wide association studies now direct animal experiments in order to explore genetic variants common in Homo sapiens. However, even these approaches must utilize animal studies for validation of function and understanding of mechanisms. Animal research currently focuses on genetic expression profiles since the majority of variants occur in non-coding regions, implying regulatory divergences. Moving forward, it will be important in both human and animal research to define the phenotypes of hearing loss and tinnitus, as well as exposure parameters, in order to extricate genes related to acoustic trauma versus those related to aging. It has become clear that common disorders like acoustic trauma are influenced by large numbers of genes, each with small effects, which cumulatively lead to susceptibility to a disorder. A polygenic risk score, which aggregates these small effect sizes of multiple genes, may offer a more accurate description of risk for NIHL and/or tinnitus.


Asunto(s)
Predisposición Genética a la Enfermedad , Pérdida Auditiva Provocada por Ruido/genética , Acúfeno/genética , Animales , Estudio de Asociación del Genoma Completo/métodos , Genómica/métodos , Humanos , Ratones , Herencia Multifactorial , Sitios de Carácter Cuantitativo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA