Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.984
Filtrar
1.
PLoS Biol ; 22(6): e3002665, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38935589

RESUMEN

Loss of synapses between spiral ganglion neurons and inner hair cells (IHC synaptopathy) leads to an auditory neuropathy called hidden hearing loss (HHL) characterized by normal auditory thresholds but reduced amplitude of sound-evoked auditory potentials. It has been proposed that synaptopathy and HHL result in poor performance in challenging hearing tasks despite a normal audiogram. However, this has only been tested in animals after exposure to noise or ototoxic drugs, which can cause deficits beyond synaptopathy. Furthermore, the impact of supernumerary synapses on auditory processing has not been evaluated. Here, we studied mice in which IHC synapse counts were increased or decreased by altering neurotrophin 3 (Ntf3) expression in IHC supporting cells. As we previously showed, postnatal Ntf3 knockdown or overexpression reduces or increases, respectively, IHC synapse density and suprathreshold amplitude of sound-evoked auditory potentials without changing cochlear thresholds. We now show that IHC synapse density does not influence the magnitude of the acoustic startle reflex or its prepulse inhibition. In contrast, gap-prepulse inhibition, a behavioral test for auditory temporal processing, is reduced or enhanced according to Ntf3 expression levels. These results indicate that IHC synaptopathy causes temporal processing deficits predicted in HHL. Furthermore, the improvement in temporal acuity achieved by increasing Ntf3 expression and synapse density suggests a therapeutic strategy for improving hearing in noise for individuals with synaptopathy of various etiologies.


Asunto(s)
Células Ciliadas Auditivas Internas , Neurotrofina 3 , Sinapsis , Animales , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patología , Sinapsis/metabolismo , Sinapsis/fisiología , Neurotrofina 3/metabolismo , Neurotrofina 3/genética , Ratones , Umbral Auditivo , Potenciales Evocados Auditivos/fisiología , Reflejo de Sobresalto/fisiología , Percepción Auditiva/fisiología , Ganglio Espiral de la Cóclea/metabolismo , Femenino , Masculino , Pérdida de Audición Oculta
2.
Hear Res ; 447: 109024, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38735179

RESUMEN

Delayed loss of residual acoustic hearing after cochlear implantation is a common but poorly understood phenomenon due to the scarcity of relevant temporal bone tissues. Prior histopathological analysis of one case of post-implantation hearing loss suggested there were no interaural differences in hair cell or neural degeneration to explain the profound loss of low-frequency hearing on the implanted side (Quesnel et al., 2016) and attributed the threshold elevation to neo-ossification and fibrosis around the implant. Here we re-evaluated the histopathology in this case, applying immunostaining and improved microscopic techniques for differentiating surviving hair cells from supporting cells. The new analysis revealed dramatic interaural differences, with a > 80 % loss of inner hair cells in the cochlear apex on the implanted side, which can account for the post-implantation loss of residual hearing. Apical degeneration of the stria further contributed to threshold elevation on the implanted side. In contrast, spiral ganglion cell survival was reduced in the region of the electrode on the implanted side, but apical counts in the two ears were similar to that seen in age-matched unimplanted control ears. Almost none of the surviving auditory neurons retained peripheral axons throughout the basal half of the cochlea. Relevance to cochlear implant performance is discussed.


Asunto(s)
Umbral Auditivo , Implantación Coclear , Implantes Cocleares , Ganglio Espiral de la Cóclea , Implantación Coclear/instrumentación , Implantación Coclear/efectos adversos , Humanos , Ganglio Espiral de la Cóclea/patología , Ganglio Espiral de la Cóclea/fisiopatología , Células Ciliadas Auditivas Internas/patología , Factores de Tiempo , Supervivencia Celular , Masculino , Audición , Pérdida Auditiva/fisiopatología , Pérdida Auditiva/patología , Pérdida Auditiva/cirugía , Pérdida Auditiva/etiología , Femenino , Células Ciliadas Auditivas/patología , Anciano , Degeneración Nerviosa , Persona de Mediana Edad , Hueso Temporal/patología , Hueso Temporal/cirugía
3.
Int J Mol Sci ; 25(10)2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38791192

RESUMEN

The synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs) are the most vulnerable structures in the noise-exposed cochlea. Cochlear synaptopathy results from the disruption of these synapses following noise exposure and is considered the main cause of poor speech understanding in noisy environments, even when audiogram results are normal. Cochlear synaptopathy leads to the degeneration of SGNs if damaged IHC-SGN synapses are not promptly recovered. Oxidative stress plays a central role in the pathogenesis of cochlear synaptopathy. C-Phycocyanin (C-PC) has antioxidant and anti-inflammatory activities and is widely utilized in the food and drug industry. However, the effect of the C-PC on noise-induced cochlear damage is unknown. We first investigated the therapeutic effect of C-PC on noise-induced cochlear synaptopathy. In vitro experiments revealed that C-PC reduced the H2O2-induced generation of reactive oxygen species in HEI-OC1 auditory cells. H2O2-induced cytotoxicity in HEI-OC1 cells was reduced with C-PC treatment. After white noise exposure for 3 h at a sound pressure of 118 dB, the guinea pigs intratympanically administered 5 µg/mL C-PC exhibited greater wave I amplitudes in the auditory brainstem response, more IHC synaptic ribbons and more IHC-SGN synapses according to microscopic analysis than the saline-treated guinea pigs. Furthermore, the group treated with C-PC had less intense 4-hydroxynonenal and intercellular adhesion molecule-1 staining in the cochlea compared with the saline group. Our results suggest that C-PC improves cochlear synaptopathy by inhibiting noise-induced oxidative stress and the inflammatory response in the cochlea.


Asunto(s)
Cóclea , Molécula 1 de Adhesión Intercelular , Ruido , Estrés Oxidativo , Ficocianina , Sinapsis , Animales , Estrés Oxidativo/efectos de los fármacos , Cobayas , Ficocianina/farmacología , Ficocianina/uso terapéutico , Cóclea/metabolismo , Cóclea/efectos de los fármacos , Cóclea/patología , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Ruido/efectos adversos , Molécula 1 de Adhesión Intercelular/metabolismo , Pérdida Auditiva Provocada por Ruido/tratamiento farmacológico , Pérdida Auditiva Provocada por Ruido/metabolismo , Pérdida Auditiva Provocada por Ruido/patología , Especies Reactivas de Oxígeno/metabolismo , Masculino , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/metabolismo , Ganglio Espiral de la Cóclea/patología , Peróxido de Hidrógeno/metabolismo , Células Ciliadas Auditivas Internas/efectos de los fármacos , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patología , Antioxidantes/farmacología , Línea Celular , Pérdida de Audición Oculta
4.
Acta Otolaryngol ; 144(3): 198-206, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38662892

RESUMEN

BACKGROUND: Recycling of synaptic vesicles plays an important role in vesicle pool replenishment, neurotransmitter release and synaptic plasticity. Clathrin-mediated endocytosis (CME) is considered to be the main mechanism for synaptic vesicle replenishment. AP-2 (adaptor-related protein complex 2) and myosin Ⅵ are known as key proteins that regulate the structure and dynamics of CME. OBJECTIVE: This study aims to reveal the spatiotemporal expression of AP-2/myosin Ⅵ in inner hair cells (IHCs) of the mouse cochlea and its correlation with auditory function. MATERIAL AND METHODS: Immunofluorescence was used to detect the localization and expression of AP-2 and myosin Ⅵ in cochlear hair cells (HCs) of CBA/CaJ mice of various ages. qRT-PCR was used to verify the differential expression of AP-2 and myosin Ⅵ mRNA in the mouse cochlea, and ABR tests were administered to mice of various ages. A preliminary analysis of the correlation between AP-2/myosin Ⅵ levels and auditory function was conducted. RESULTS: AP-2 was located in the cytoplasmic region of IHCs and was mainly expressed in the basal region of IHCs and the area near ribbon synapses, while myosin Ⅵ was expressed in the cytoplasmic region of IHCs and OHCs. Furthermore, AP-2 and myosin Ⅵ were not significant detected in the cochleae of P7 mice; the expression level reached a peak at P35 and then decreased significantly with age. The expression patterns and expression levels of AP-2 and myosin Ⅵ in the cochleae of the mice were consistent with the development of the auditory system. CONCLUSIONS AND SIGNIFICANCE: AP-2 and myosin Ⅵ protein expression may differ in mice of different ages, and this variation probably leads to a difference in the efficiency in CME; it may also cause a defect in IHC function.


Asunto(s)
Células Ciliadas Auditivas Internas , Ratones Endogámicos CBA , Animales , Células Ciliadas Auditivas Internas/metabolismo , Ratones , Complejo 2 de Proteína Adaptadora/metabolismo , Complejo 2 de Proteína Adaptadora/genética , Potenciales Evocados Auditivos del Tronco Encefálico , Miosina Tipo IIB no Muscular/metabolismo , Miosina Tipo IIB no Muscular/genética , Cóclea/metabolismo
5.
J Chem Neuroanat ; 137: 102417, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38570170

RESUMEN

OBJECTIVE: The distribution and role of NMDA receptors is unclear in the afferent signaling complex of the cochlea. The present study aimed to examine the distribution of NMDA receptors in cochlear afferent signaling complex of the adult mouse, and their relationship with ribbon synapses of inner hair cells (IHCs) and GABAergic efferent terminals of the lateral olivocochlear (LOC). METHODS: Immunofluorescence staining in combination with confocal microscopy was used to investigate the distribution of glutamatergic NMDA and AMPA receptors in afferent terminals of SGNs, and their relationship with ribbon synapses of IHCs and GABAergic efferent terminals of LOC. RESULTS: Terminals with AMPA receptors along with Ribbons of IHC formed afferent synapses in the basal pole of IHCs, and those with NMDA receptors were mainly distributed longitudinally in the IHCs nuclei region. Significant difference was found in the distribution of NMDA and AMPA receptors in IHC afferent signaling complex (P<0.05). Some GABAergic terminals colocalized with NMDA receptors at the IHC nucleus region (P>0.05). CONCLUSION: There is significant difference in the distribution of NMDA and AMPA receptors in cochlear afferent signaling complex. NMDA receptors are present in the extra-synaptic region of ribbon synapses of IHCs, and they are related to GABA efferent terminals of the afferent signaling complex.


Asunto(s)
Células Ciliadas Auditivas Internas , Receptores AMPA , Receptores de N-Metil-D-Aspartato , Sinapsis , Animales , Células Ciliadas Auditivas Internas/metabolismo , Ratones , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Receptores AMPA/metabolismo , Cóclea/metabolismo , Masculino
6.
J Neurosci ; 44(23)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38688721

RESUMEN

The mouse auditory organ cochlea contains two types of sound receptors: inner hair cells (IHCs) and outer hair cells (OHCs). Tbx2 is expressed in IHCs but repressed in OHCs, and neonatal OHCs that misexpress Tbx2 transdifferentiate into IHC-like cells. However, the extent of this switch from OHCs to IHC-like cells and the underlying molecular mechanism remain poorly understood. Furthermore, whether Tbx2 can transform fully mature adult OHCs into IHC-like cells is unknown. Here, our single-cell transcriptomic analysis revealed that in neonatal OHCs misexpressing Tbx2, 85.6% of IHC genes, including Slc17a8, are upregulated, but only 38.6% of OHC genes, including Ikzf2 and Slc26a5, are downregulated. This suggests that Tbx2 cannot fully reprogram neonatal OHCs into IHCs. Moreover, Tbx2 also failed to completely reprogram cochlear progenitors into IHCs. Lastly, restoring Ikzf2 expression alleviated the abnormalities detected in Tbx2+ OHCs, which supports the notion that Ikzf2 repression by Tbx2 contributes to the transdifferentiation of OHCs into IHC-like cells. Our study evaluates the effects of ectopic Tbx2 expression on OHC lineage development at distinct stages of either male or female mice and provides molecular insights into how Tbx2 disrupts the gene expression profile of OHCs. This research also lays the groundwork for future studies on OHC regeneration.


Asunto(s)
Células Ciliadas Auditivas Internas , Células Ciliadas Auditivas Externas , Proteínas de Dominio T Box , Animales , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/genética , Ratones , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Femenino , Animales Recién Nacidos , Transdiferenciación Celular/fisiología , Transdiferenciación Celular/genética , Masculino , Cóclea/metabolismo , Cóclea/citología , Ratones Endogámicos C57BL
7.
Cell Rep ; 43(4): 114025, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38564333

RESUMEN

Type I spiral ganglion neurons (SGNs) convey sound information to the central auditory pathway by forming synapses with inner hair cells (IHCs) in the mammalian cochlea. The molecular mechanisms regulating the formation of the post-synaptic density (PSD) in the SGN afferent terminals are still unclear. Here, we demonstrate that brain-specific angiogenesis inhibitor 1 (BAI1) is required for the clustering of AMPA receptors GluR2-4 (glutamate receptors 2-4) at the PSD. Adult Bai1-deficient mice have functional IHCs but fail to transmit information to the SGNs, leading to highly raised hearing thresholds. Despite the almost complete absence of AMPA receptor subunits, the SGN fibers innervating the IHCs do not degenerate. Furthermore, we show that AMPA receptors are still expressed in the cochlea of Bai1-deficient mice, highlighting a role for BAI1 in trafficking or anchoring GluR2-4 to the PSDs. These findings identify molecular and functional mechanisms required for sound encoding at cochlear ribbon synapses.


Asunto(s)
Cóclea , Audición , Densidad Postsináptica , Receptores AMPA , Receptores Acoplados a Proteínas G , Ganglio Espiral de la Cóclea , Animales , Receptores AMPA/metabolismo , Ratones , Ganglio Espiral de la Cóclea/metabolismo , Audición/fisiología , Cóclea/metabolismo , Densidad Postsináptica/metabolismo , Ratones Noqueados , Células Ciliadas Auditivas Internas/metabolismo , Ratones Endogámicos C57BL , Sinapsis/metabolismo
8.
Hum Mol Genet ; 33(10): 905-918, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38449065

RESUMEN

Mutations in AIFM1, encoding for apoptosis-inducing factor (AIF), cause AUNX1, an X-linked neurologic disorder with late-onset auditory neuropathy (AN) and peripheral neuropathy. Despite significant research on AIF, there are limited animal models with the disrupted AIFM1 representing the corresponding phenotype of human AUNX1, characterized by late-onset hearing loss and impaired auditory pathways. Here, we generated an Aifm1 p.R450Q knock-in mouse model (KI) based on the human AIFM1 p.R451Q mutation. Hemizygote KI male mice exhibited progressive hearing loss from P30 onward, with greater severity at P60 and stabilization until P210. Additionally, muscle atrophy was observed at P210. These phenotypic changes were accompanied by a gradual reduction in the number of spiral ganglion neuron cells (SGNs) at P30 and ribbons at P60, which coincided with the translocation of AIF into the nucleus starting from P21 and P30, respectively. The SGNs of KI mice at P210 displayed loss of cytomembrane integrity, abnormal nuclear morphology, and dendritic and axonal demyelination. Furthermore, the inner hair cells and myelin sheath displayed abnormal mitochondrial morphology, while fibroblasts from KI mice showed impaired mitochondrial function. In conclusion, we successfully generated a mouse model recapitulating AUNX1. Our findings indicate that disruption of Aifm1 induced the nuclear translocation of AIF, resulting in the impairment in the auditory pathway.


Asunto(s)
Factor Inductor de la Apoptosis , Modelos Animales de Enfermedad , Pérdida Auditiva , Animales , Humanos , Masculino , Ratones , Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/genética , Técnicas de Sustitución del Gen , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patología , Pérdida Auditiva/genética , Pérdida Auditiva/patología , Pérdida Auditiva/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patología , Atrofia Muscular/metabolismo , Mutación , Transporte de Proteínas , Ganglio Espiral de la Cóclea/metabolismo , Ganglio Espiral de la Cóclea/patología
9.
Int J Mol Sci ; 25(5)2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38473985

RESUMEN

In mammalian hearing, type-I afferent auditory nerve fibers comprise the basis of the afferent auditory pathway. They are connected to inner hair cells of the cochlea via specialized ribbon synapses. Auditory nerve fibers of different physiological types differ subtly in their synaptic location and morphology. Low-spontaneous-rate auditory nerve fibers typically connect on the modiolar side of the inner hair cell, while high-spontaneous-rate fibers are typically found on the pillar side. In aging and noise-damaged ears, this fine-tuned balance between auditory nerve fiber populations can be disrupted and the functional consequences are currently unclear. Here, using immunofluorescent labeling of presynaptic ribbons and postsynaptic glutamate receptor patches, we investigated changes in synaptic morphology at three different tonotopic locations along the cochlea of aging gerbils compared to those of young adults. Quiet-aged gerbils showed about 20% loss of afferent ribbon synapses. While the loss was random at apical, low-frequency cochlear locations, at the basal, high-frequency location it almost exclusively affected the modiolar-located synapses. The subtle differences in volumes of pre- and postsynaptic elements located on the inner hair cell's modiolar versus pillar side were unaffected by age. This is consistent with known physiology and suggests a predominant, age-related loss in the low-spontaneous-rate auditory nerve population in the cochlear base, but not the apex.


Asunto(s)
Cóclea , Sinapsis , Animales , Gerbillinae , Cóclea/metabolismo , Sinapsis/metabolismo , Nervio Coclear/metabolismo , Células Ciliadas Auditivas Internas/metabolismo
10.
Elife ; 132024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38334748

RESUMEN

Two calcium-binding proteins, CaBP1 and CaBP2, cooperate to keep calcium channels in the hair cells of the inner ear open.


Asunto(s)
Calcio , Células Ciliadas Auditivas , Calcio/metabolismo , Células Ciliadas Auditivas/metabolismo , Canales de Calcio/metabolismo , Calcio de la Dieta , Células Ciliadas Auditivas Internas/metabolismo , Proteínas de Unión al Calcio/metabolismo
11.
Hear Res ; 443: 108966, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38310710

RESUMEN

The nonlinearities of the inner ear are often considered to be obstacles that the central nervous system has to overcome to decode neural responses to sounds. This review describes how peripheral nonlinearities, such as saturation of the inner-hair-cell response and of the IHC-auditory-nerve synapse, are instead beneficial to the neural encoding of complex sounds such as speech. These nonlinearities set up contrast in the depth of neural-fluctuations in auditory-nerve responses along the tonotopic axis, referred to here as neural fluctuation contrast (NFC). Physiological support for the NFC coding hypothesis is reviewed, and predictions of several psychophysical phenomena, including masked detection and speech intelligibility, are presented. Lastly, a framework based on the NFC code for understanding how the medial olivocochlear (MOC) efferent system contributes to the coding of complex sounds is presented. By modulating cochlear gain control in response to both sound energy and fluctuations in neural responses, the MOC system is hypothesized to function not as a simple feedback gain-control device, but rather as a mechanism for enhancing NFC along the tonotopic axis, enabling robust encoding of complex sounds across a wide range of sound levels and in the presence of background noise. Effects of sensorineural hearing loss on the NFC code and on the MOC feedback system are presented and discussed.


Asunto(s)
Cóclea , Pérdida Auditiva Sensorineural , Humanos , Cóclea/fisiología , Ruido/efectos adversos , Nervio Coclear , Células Ciliadas Auditivas Internas/fisiología
12.
Proc Natl Acad Sci U S A ; 121(10): e2309656121, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38408254

RESUMEN

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.


Asunto(s)
Sordera , Pérdida Auditiva , Ratones , Animales , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Células Ciliadas Auditivas/fisiología , Sordera/genética , Células Ciliadas Auditivas Internas/metabolismo , Cadherinas/metabolismo , Exones/genética
13.
Nat Commun ; 15(1): 526, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38228630

RESUMEN

The inner ear is the hub where hair cells (HCs) transduce sound, gravity, and head acceleration stimuli to the brain. Hearing and balance rely on mechanosensation, the fastest sensory signals transmitted to the brain. The mechanoelectrical transducer (MET) channel is the entryway for the sound-balance-brain interface, but the channel-complex composition is not entirely known. Here, we report that the mouse utilizes Piezo1 (Pz1) and Piezo2 (Pz2) isoforms as MET-complex components. The Pz channels, expressed in HC stereocilia, and cell lines are co-localized and co-assembled with MET complex partners. Mice expressing non-functional Pz1 and Pz2 at the ROSA26 locus have impaired auditory and vestibular traits that can only be explained if the Pzs are integral to the MET complex. We suggest that Pz subunits constitute part of the MET complex and that interactions with other MET complex components yield functional MET units to generate HC MET currents.


Asunto(s)
Oído Interno , Células Ciliadas Auditivas Internas , Animales , Ratones , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas/metabolismo , Estereocilios/metabolismo , Oído Interno/metabolismo , Audición , Mecanotransducción Celular , Mamíferos/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo
14.
J Neurosci ; 44(4)2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38050104

RESUMEN

Outer hair cells (OHCs) of the organ of Corti (OoC), acting as bidirectional cellular mechanoelectrical transducers, generate, receive, and exchange forces with other major elements of the cochlear partition, including the sensory inner hair cells (IHCs). Force exchange is mediated via a supporting cell scaffold, including Deiters' (DC) and outer pillar cells (OPC), to enable the sensitivity and exquisite frequency selectivity of the mammalian cochlea and to transmit its responses to the auditory nerve. To selectively activate DCs and OPCs in male and female mice, we conditionally expressed in them a hyperpolarizing halorhodopsin (HOP), a light-gated inward chloride ion pump, and measured extracellular receptor potentials (ERPs) and their DC component (ERPDCs) from the cortilymph, which fills the OoC fluid spaces, and compared the responses with similar potentials from HOP-/- littermates. The compound action potentials (CAP) of the auditory nerve were measured as an indication of IHC activity and transmission of cochlear responses to the CNS. HOP light-activated hyperpolarization of DCs and OPCs suppressed cochlear amplification through changing the timing of its feedback, altered basilar membrane (BM) responses to tones at all measured levels and frequencies, and reduced IHC excitation. HOP activation findings reported here complement recent studies that revealed channelrhodopsin activation depolarized DCs and OPCs and effectively bypassed, rather than blocked, the control of OHC mechanical and electrical responses to sound and their contribution to timed and directed electromechanical feedback to the mammalian cochlea. Moreover, our findings identify DCs and OPCs as potential targets for the treatment of noise-induced hearing loss.


Asunto(s)
Células Ciliadas Auditivas Externas , Células Ciliadas Vestibulares , Femenino , Masculino , Ratones , Animales , Células Ciliadas Auditivas Externas/fisiología , Optogenética , Cóclea/fisiología , Células Ciliadas Auditivas Internas/fisiología , Órgano Espiral/fisiología , Mamíferos
15.
Neurosci Bull ; 40(1): 113-126, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37787875

RESUMEN

Hearing loss has become increasingly prevalent and causes considerable disability, thus gravely burdening the global economy. Irreversible loss of hair cells is a main cause of sensorineural hearing loss, and currently, the only relatively effective clinical treatments are limited to digital hearing equipment like cochlear implants and hearing aids, but these are of limited benefit in patients. It is therefore urgent to understand the mechanisms of damage repair in order to develop new neuroprotective strategies. At present, how to promote the regeneration of functional hair cells is a key scientific question in the field of hearing research. Multiple signaling pathways and transcriptional factors trigger the activation of hair cell progenitors and ensure the maturation of newborn hair cells, and in this article, we first review the principal mechanisms underlying hair cell reproduction. We then further discuss therapeutic strategies involving the co-regulation of multiple signaling pathways in order to induce effective functional hair cell regeneration after degeneration, and we summarize current achievements in hair cell regeneration. Lastly, we discuss potential future approaches, such as small molecule drugs and gene therapy, which might be applied for regenerating functional hair cells in the clinic.


Asunto(s)
Oído Interno , Células Ciliadas Auditivas Internas , Recién Nacido , Humanos , Células Ciliadas Auditivas Internas/fisiología , Oído Interno/fisiología , Células Ciliadas Auditivas/fisiología , Regeneración/genética , Células Madre
16.
Neuroscience Bulletin ; (6): 113-126, 2024.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-1010674

RESUMEN

Hearing loss has become increasingly prevalent and causes considerable disability, thus gravely burdening the global economy. Irreversible loss of hair cells is a main cause of sensorineural hearing loss, and currently, the only relatively effective clinical treatments are limited to digital hearing equipment like cochlear implants and hearing aids, but these are of limited benefit in patients. It is therefore urgent to understand the mechanisms of damage repair in order to develop new neuroprotective strategies. At present, how to promote the regeneration of functional hair cells is a key scientific question in the field of hearing research. Multiple signaling pathways and transcriptional factors trigger the activation of hair cell progenitors and ensure the maturation of newborn hair cells, and in this article, we first review the principal mechanisms underlying hair cell reproduction. We then further discuss therapeutic strategies involving the co-regulation of multiple signaling pathways in order to induce effective functional hair cell regeneration after degeneration, and we summarize current achievements in hair cell regeneration. Lastly, we discuss potential future approaches, such as small molecule drugs and gene therapy, which might be applied for regenerating functional hair cells in the clinic.


Asunto(s)
Recién Nacido , Humanos , Células Ciliadas Auditivas Internas/fisiología , Oído Interno/fisiología , Células Ciliadas Auditivas/fisiología , Regeneración/genética , Células Madre
17.
Biochem Biophys Res Commun ; 693: 149396, 2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38118309

RESUMEN

Zinc plays a vital role in our metabolism, encompassing antioxidant regulation, immune response, and auditory function. Several studies have reported that zinc levels correlate with hearing loss. We have previously demonstrated that the auditory brainstem response (ABR) threshold increased in mice fed a zinc-deficient diet. However, the effects of zinc deficiency on hearing were not fully elucidated. The present study investigated whether zinc deficiency affects hearing in association with neuronal components or cochlear structures. CBA/N mice were fed a normal or zinc-deficient diet for 8 weeks and assessed for ABR and distortion product otoacoustic emissions (DPOAE). The cochlear sections were stained with hematoxylin and eosin solution. Also, we observed the expression of synaptic ribbons, neurofilaments, and alpha-synuclein (α-Syn). The 8-week zinc-deficient diet mice had an elevated ABR threshold but no changed DPOAE threshold or cochlear structures. A reduced number of synaptic ribbons of inner hair cells (IHCs) and impaired efferent nerve fibers were observed in the zinc-deficient diet mice. The number of outer hair cells (OHCs) and expression of α-Syn remained unchanged. Our results suggest that zinc-mediated hearing loss is associated with the loss of neuronal components of IHCs.


Asunto(s)
Sordera , Pérdida Auditiva , Animales , Ratones , Células Ciliadas Auditivas Internas/metabolismo , Ratones Endogámicos CBA , Cóclea/metabolismo , Sinapsis/metabolismo , Sordera/metabolismo , Zinc/metabolismo , Potenciales Evocados Auditivos del Tronco Encefálico , Umbral Auditivo
18.
Toxicol Lett ; 391: 86-99, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38101494

RESUMEN

Ototoxicity is a major side effect of aminoglycosides, which can cause irreversible hearing loss. Previous studies on aminoglycoside-induced ototoxicity have primarily focused on the loss of sensory hair cells. Recent investigations have revealed that aminoglycosides can also lead to the loss of ribbon synapses in inner hair cells (IHCs). However, the functional implications of ribbon synapse loss and the underlying mechanisms remain unclear. In this study, we intraperitoneally injected C57BL/6 J mice with 300 mg/kg gentamicin once daily for 3, 10, and 20 days. Then, we performed immunofluorescence staining, patch-clamp recording, proteomics analysis and western blotting to characterize the changes in ribbon synapses in IHCs and the associated mechanisms. After gentamicin treatment, the auditory brainstem response (ABR) threshold was elevated, and the ABR wave I amplitude was decreased. We also observed loss of ribbon synapses in IHCs. Interestingly, ribbon synapse loss occurred on both the modiolar and pillar sides of IHCs. Whole-cell patch-clamp recordings in IHCs revealed a reduction in the calcium current amplitude, along with a shifted half-activation voltage and altered calcium voltage dependency. Moreover, exocytosis of IHCs was reduced, consistent with the reduction in the ABR wave I amplitude. Through proteomic analysis, western blotting, and immunofluorescence staining, we found that gentamicin treatment resulted in downregulation of myosin VI, a protein crucial for synaptic vesicle recycling and replenishment in IHCs. Furthermore, we evaluated the kinetics of endocytosis and found a significant reduction in IHC exocytosis, possibly reflecting the impact of myosin VI downregulation on synaptic vesicle recycling. In summary, our findings demonstrate that gentamicin treatment leads to synaptic dysfunction in IHCs, highlighting the important role of myosin VI downregulation in gentamicin-induced synaptic damage.


Asunto(s)
Células Ciliadas Auditivas Internas , Ototoxicidad , Animales , Ratones , Calcio/metabolismo , Proteómica , Ratones Endogámicos C57BL , Sinapsis , Gentamicinas/toxicidad , Antibacterianos/toxicidad , Aminoglicósidos/metabolismo , Aminoglicósidos/farmacología , Cóclea
19.
Mol Cell Proteomics ; 23(2): 100704, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38128648

RESUMEN

In the ear, inner hair cells (IHCs) employ sophisticated glutamatergic ribbon synapses with afferent neurons to transmit auditory information to the brain. The presynaptic machinery responsible for neurotransmitter release in IHC synapses includes proteins such as the multi-C2-domain protein otoferlin and the vesicular glutamate transporter 3 (VGluT3). Yet, much of this likely unique molecular machinery remains to be deciphered. The scarcity of material has so far hampered biochemical studies which require large amounts of purified samples. We developed a subcellular fractionation workflow combined with immunoisolation of VGluT3-containing membrane vesicles, allowing for the enrichment of glutamatergic organelles that are likely dominated by synaptic vesicles (SVs) of IHCs. We have characterized their protein composition in mice before and after hearing onset using mass spectrometry and confocal imaging and provide a fully annotated proteome with hitherto unidentified proteins. Despite the prevalence of IHC marker proteins across IHC maturation, the profiles of trafficking proteins differed markedly before and after hearing onset. Among the proteins enriched after hearing onset were VAMP-7, syntaxin-7, syntaxin-8, syntaxin-12/13, SCAMP1, V-ATPase, SV2, and PKCα. Our study provides an inventory of the machinery associated with synaptic vesicle-mediated trafficking and presynaptic activity at IHC ribbon synapses and serves as a foundation for future functional studies.


Asunto(s)
Células Ciliadas Auditivas Internas , Proteómica , Ratones , Animales , Células Ciliadas Auditivas Internas/metabolismo , Sinapsis/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas de la Membrana/metabolismo
20.
Development ; 150(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38078650

RESUMEN

Cochlear inner hair cells (IHCs) are primary sound receptors, and are therefore a target for developing treatments for hearing impairment. IHC regeneration in vivo has been widely attempted, although not yet in the IHC-damaged cochlea. Moreover, the extent to which new IHCs resemble wild-type IHCs remains unclear, as is the ability of new IHCs to improve hearing. Here, we have developed an in vivo mouse model wherein wild-type IHCs were pre-damaged and nonsensory supporting cells were transformed into IHCs by ectopically expressing Atoh1 transiently and Tbx2 permanently. Notably, the new IHCs expressed the functional marker vGlut3 and presented similar transcriptomic and electrophysiological properties to wild-type IHCs. Furthermore, the formation efficiency and maturity of new IHCs were higher than those previously reported, although marked hearing improvement was not achieved, at least partly due to defective mechanoelectrical transduction (MET) in new IHCs. Thus, we have successfully regenerated new IHCs resembling wild-type IHCs in many respects in the damaged cochlea. Our findings suggest that the defective MET is a critical barrier that prevents the restoration of hearing capacity and should thus facilitate future IHC regeneration studies.


Asunto(s)
Células Ciliadas Vestibulares , Pérdida Auditiva , Ratones , Animales , Células Ciliadas Auditivas Internas , Cóclea/fisiología , Pérdida Auditiva/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...