Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
1.
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
2.
Neurosci Lett ; 772: 136493, 2022 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-35114332

RESUMEN

Autophagy plays a pathogenic role in neurodegenerative disease. However, the involvement of autophagy in the pathogenesis of age-related hearing loss (ARHL) remains obscure. Naturally aged C57BL/6J mice were used to identify the role of autophagy in ARHL, and rapamycin, a mammalian target of rapamycin (mTOR) inhibitor, was administered for 34 weeks to explore the potential therapeutic effect of rapamycin in ARHL. We found that the number of autophagosomes and the expression of microtubule-associated protein 1 light chain 3B (LC3B) decreased as the mice aged. The expression of autophagy-related (Atg) proteins, including Beclin1 and Atg5, and the ratio of LC3-II/I was reduced in aged mice, while mTOR activity in aged mice gradually increased. Rapamycin improved the auditory brainstem response (ABR) threshold (at 8, 12, and 24 kHz). Further exploration demonstrated that spiral ganglion neuron (SGN) density was enhanced in response to administration of rapamycin. The rate of apoptosis in the basal turn SGNs was decreased, whereas autophagy activity was increased in the experimental group. Meanwhile, mTOR activity in the experimental group was decreased. Our findings indicate that age-related deficiency in autophagy may lead to increased apoptosis of aged SGNs. Rapamycin enhances autophagy of SGNs by inhibiting mTOR activation, resulting in amelioration of ARHL. Therapeutic strategy targeting autophagy may provide a potential approach for treating ARHL.


Asunto(s)
Envejecimiento/patología , Autofagia , Pérdida Auditiva/tratamiento farmacológico , Sirolimus/farmacología , Ganglio Espiral de la Cóclea/efectos de los fármacos , Envejecimiento/metabolismo , Animales , Proteína 5 Relacionada con la Autofagia/metabolismo , Beclina-1/metabolismo , Potenciales Evocados Auditivos del Tronco Encefálico , Pérdida Auditiva/metabolismo , Pérdida Auditiva/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Sirolimus/uso terapéutico , Ganglio Espiral de la Cóclea/metabolismo , Ganglio Espiral de la Cóclea/fisiopatología , Serina-Treonina Quinasas TOR/metabolismo
3.
J Neurophysiol ; 126(6): 2027-2038, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34788179

RESUMEN

Cochlear synaptopathy is the noise-induced or age-related loss of ribbon synapses between inner hair cells (IHCs) and auditory-nerve fibers (ANFs), first reported in CBA/CaJ mice. Recordings from single ANFs in anesthetized, noise-exposed guinea pigs suggested that neurons with low spontaneous rates (SRs) and high thresholds are more vulnerable than low-threshold, high-SR fibers. However, there is extensive postexposure regeneration of ANFs in guinea pigs but not in mice. Here, we exposed CBA/CaJ mice to octave-band noise and recorded sound-evoked and spontaneous activity from single ANFs at least 2 wk later. Confocal analysis of cochleae immunostained for pre- and postsynaptic markers confirmed the expected loss of 40%-50% of ANF synapses in the basal half of the cochlea; however, our data were not consistent with a selective loss of low-SR fibers. Rather they suggested a loss of both SR groups in synaptopathic regions. Single-fiber thresholds and frequency tuning recovered to pre-exposure levels; however, response to tone bursts showed increased peak and steady-state firing rates, as well as decreased jitter in first-spike latencies. This apparent gain-of-function increased the robustness of tone-burst responses in the presence of continuous masking noise. This study suggests that the nature of noise-induced synaptic damage varies between different species and that, in mouse, the noise-induced hyperexcitability seen in central auditory circuits is also observed at the level of the auditory nerve.NEW & NOTEWORTHY Noise-induced damage to synapses between inner hair cells and auditory-nerve fibers (ANFs) can occur without permanent hair cell damage, resulting in pathophysiology that "hides" behind normal thresholds. Prior single-fiber neurophysiology in guinea pig suggested that noise selectively targets high-threshold ANFs. Here, we show that the lingering pathophysiology differs in mouse, with both ANF groups affected and a paradoxical gain-of-function in surviving low-threshold fibers, including increased onset rate, decreased onset jitter, and reduced maskability.


Asunto(s)
Enfermedades Cocleares/fisiopatología , Nervio Coclear/fisiopatología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Ganglio Espiral de la Cóclea/fisiopatología , Sinapsis/patología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos CBA
4.
Cell Rep ; 36(13): 109758, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34592158

RESUMEN

Noise-induced hearing loss (NIHL) results from a complex interplay of damage to the sensory cells of the inner ear, dysfunction of its lateral wall, axonal retraction of type 1C spiral ganglion neurons, and activation of the immune response. We use RiboTag and single-cell RNA sequencing to survey the cell-type-specific molecular landscape of the mouse inner ear before and after noise trauma. We identify induction of the transcription factors STAT3 and IRF7 and immune-related genes across all cell-types. Yet, cell-type-specific transcriptomic changes dominate the response. The ATF3/ATF4 stress-response pathway is robustly induced in the type 1A noise-resilient neurons, potassium transport genes are downregulated in the lateral wall, mRNA metabolism genes are downregulated in outer hair cells, and deafness-associated genes are downregulated in most cell types. This transcriptomic resource is available via the Gene Expression Analysis Resource (gEAR; https://umgear.org/NIHL) and provides a blueprint for the rational development of drugs to prevent and treat NIHL.


Asunto(s)
Oído Interno/metabolismo , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva Provocada por Ruido/metabolismo , Pérdida Auditiva Provocada por Ruido/fisiopatología , Ganglio Espiral de la Cóclea/metabolismo , Animales , Cóclea/metabolismo , Cóclea/fisiopatología , Oído Interno/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Pérdida Auditiva Provocada por Ruido/genética , Ratones , Neuronas/metabolismo , Ruido , Ganglio Espiral de la Cóclea/citología , Ganglio Espiral de la Cóclea/fisiopatología
5.
PLoS Comput Biol ; 17(1): e1008499, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33481777

RESUMEN

Hidden hearing loss (HHL) is an auditory neuropathy characterized by normal hearing thresholds but reduced amplitudes of the sound-evoked auditory nerve compound action potential (CAP). In animal models, HHL can be caused by moderate noise exposure or aging, which induces loss of inner hair cell (IHC) synapses. In contrast, recent evidence has shown that transient loss of cochlear Schwann cells also causes permanent auditory deficits in mice with similarities to HHL. Histological analysis of the cochlea after auditory nerve remyelination showed a permanent disruption of the myelination patterns at the heminode of type I spiral ganglion neuron (SGN) peripheral terminals, suggesting that this defect could be contributing to HHL. To shed light on the mechanisms of different HHL scenarios observed in animals and to test their impact on type I SGN activity, we constructed a reduced biophysical model for a population of SGN peripheral axons whose activity is driven by a well-accepted model of cochlear sound processing. We found that the amplitudes of simulated sound-evoked SGN CAPs are lower and have greater latencies when heminodes are disorganized, i.e. they occur at different distances from the hair cell rather than at the same distance as in the normal cochlea. These results confirm that disruption of heminode positions causes desynchronization of SGN spikes leading to a loss of temporal resolution and reduction of the sound-evoked SGN CAP. Another mechanism resulting in HHL is loss of IHC synapses, i.e., synaptopathy. For comparison, we simulated synaptopathy by removing high threshold IHC-SGN synapses and found that the amplitude of simulated sound-evoked SGN CAPs decreases while latencies remain unchanged, as has been observed in noise exposed animals. Thus, model results illuminate diverse disruptions caused by synaptopathy and demyelination on neural activity in auditory processing that contribute to HHL as observed in animal models and that can contribute to perceptual deficits induced by nerve damage in humans.


Asunto(s)
Pérdida Auditiva/fisiopatología , Vaina de Mielina , Sinapsis , Animales , Cóclea/fisiopatología , Nervio Coclear/fisiopatología , Modelos Animales de Enfermedad , Células Ciliadas Auditivas Internas/patología , Células Ciliadas Auditivas Internas/fisiología , Ratones , Modelos Neurológicos , Vaina de Mielina/patología , Vaina de Mielina/fisiología , Ganglio Espiral de la Cóclea/citología , Ganglio Espiral de la Cóclea/fisiopatología , Sinapsis/patología , Sinapsis/fisiología
6.
Otolaryngol Clin North Am ; 54(1): 189-200, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33243375

RESUMEN

Sensorineural hearing loss is caused by irreversible loss of auditory hair cells and/or neurons and is increasing in prevalence. Hair cells and neurons do not regenerate after damage, but novel regeneration therapies based on small molecule drugs, gene therapy, and cell replacement strategies offer promising therapeutic options. Endogenous and exogenous regeneration techniques are discussed in context of their feasibility for hair cell and neuron regeneration. Gene therapy and treatment of synaptopathy represent promising future therapies. Minimally invasive endoscopic ear surgery offers a viable approach to aid in delivery of pharmacologic compounds, cells, or viral vectors to the inner ear for all of these techniques.


Asunto(s)
Sistemas de Liberación de Medicamentos , Endoscopía/métodos , Pérdida Auditiva Sensorineural/terapia , Animales , Oído Interno/efectos de los fármacos , Oído Interno/fisiopatología , Terapia Genética/métodos , Células Ciliadas Auditivas Internas/patología , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Regeneración , Ganglio Espiral de la Cóclea/fisiopatología
7.
Int J Mol Sci ; 21(22)2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33198187

RESUMEN

Sensorineural deafness is caused by the loss of peripheral neural input to the auditory nerve, which may result from peripheral neural degeneration and/or a loss of inner hair cells. Provided spiral ganglion cells and their central processes are patent, cochlear implants can be used to electrically stimulate the auditory nerve to facilitate hearing in the deaf or severely hard-of-hearing. Neural degeneration is a crucial impediment to the functional success of a cochlear implant. The present, first-of-its-kind two-dimensional finite-element model investigates how the depletion of neural tissues might alter the electrically induced transmembrane potential of spiral ganglion neurons. The study suggests that even as little as 10% of neural tissue degeneration could lead to a disproportionate change in the stimulation profile of the auditory nerve. This result implies that apart from encapsulation layer formation around the cochlear implant electrode, tissue degeneration could also be an essential reason for the apparent inconsistencies in the functionality of cochlear implants.


Asunto(s)
Cóclea/fisiopatología , Nervio Coclear/fisiopatología , Degeneración Nerviosa/fisiopatología , Implantes Cocleares , Sordera/fisiopatología , Estimulación Eléctrica/métodos , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Potenciales de la Membrana/fisiología , Neuronas , Ganglio Espiral de la Cóclea/fisiopatología
8.
Neural Plast ; 2020: 8843485, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32908487

RESUMEN

Sensorineural hearing loss (SNHL) becomes an inevitable worldwide public health issue, and deafness treatment is urgently imperative; yet their current curative therapy is limited. Auditory neuropathies (AN) were proved to play a substantial role in SNHL recently, and spiral ganglion neuron (SGN) dysfunction is a dominant pathogenesis of AN. Auditory pathway is a high energy consumption system, and SGNs required sufficient mitochondria. Mitochondria are known treatment target of SNHL, but mitochondrion mechanism and pathology in SGNs are not valued. Mitochondrial dysfunction and pharmacological therapy were studied in neurodegeneration, providing new insights in mitochondrion-targeted treatment of AN. In this review, we summarized mitochondrial biological functions related to SGNs and discussed interaction between mitochondrial dysfunction and AN, as well as existing mitochondrion treatment for SNHL. Pharmaceutical exploration to protect mitochondrion dysfunction is a feasible and effective therapeutics for AN.


Asunto(s)
Pérdida Auditiva Central/fisiopatología , Pérdida Auditiva Central/terapia , Mitocondrias/fisiología , Ganglio Espiral de la Cóclea/fisiopatología , Animales , Vías Auditivas/fisiopatología , Humanos , Ratones , Neuronas/fisiología
9.
Neural Plast ; 2020: 8829660, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32802037

RESUMEN

The hair cells that reside in the cochlear sensory epithelium are the fundamental sensory structures responsible for understanding the mechanical sound waves evoked in the environment. The intense damage to these sensory structures may result in permanent hearing loss. The present strategies to rehabilitate the hearing function include either hearing aids or cochlear implants that may recover the hearing capability of deaf patients to a limited extent. Therefore, much attention has been paid on developing regenerative therapies to regenerate/replace the lost hair cells to treat the damaged cochlear sensory epithelium. The stem cell therapy is a promising approach to develop the functional hair cells and neuronal cells from endogenous and exogenous stem cell pool to recover hearing loss. In this review, we specifically discuss the potential of different kinds of stem cells that hold the potential to restore sensorineural hearing loss in mammals and comprehensively explain the current therapeutic applications of stem cells in both the human and mouse inner ear to regenerate/replace the lost hair cells and spiral ganglion neurons.


Asunto(s)
Células Ciliadas Auditivas/fisiología , Pérdida Auditiva Sensorineural/fisiopatología , Pérdida Auditiva Sensorineural/terapia , Regeneración , Trasplante de Células Madre , Animales , Humanos , Ganglio Espiral de la Cóclea/fisiopatología , Células Madre/fisiología
10.
Hear Res ; 390: 107928, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32143110

RESUMEN

Severe damage to the organ of Corti leads to degeneration of the spiral ganglion cells (SGCs) which form the auditory nerve. This degeneration starts at the level of synaptic connection of the peripheral processes (PPs) of SGCs with the cochlear hair cells. It is generally thought that from this point SGC degeneration progresses in a retrograde fashion: PPs degenerate first, followed by the SGC soma with a delay of several weeks to many months. Evidence for this course of events, both in animals and in humans, is not unambiguous, while this knowledge is important since the presence or absence of the different neural elements may greatly influence the response to electrical stimulation with a cochlear implant (CI). We therefore aimed to provide a comprehensive account of the course of SGC degeneration in the guinea pig cochlea after ototoxic treatment. Histological analysis of eighteen healthy and thirty-three deafened cochleas showed that the degeneration of SGCs and their peripheral processes was simultaneous rather than sequential. As the site of excitation for electrical stimulation with a CI may depend on the course of degeneration of the various neural elements, this finding is relevant both for understanding the electrophysiological mechanisms behind cochlear implantation and for recent efforts to induce PP resprouting for improved electrode-neural interface. Since excitation of the PPs is often thought to result in (secondary) longer-latency activity, we tested the hypothesis that having relatively many PPs produces a larger N2 peak in the electrically evoked compound action potential (eCAP); the present findings however do not support this theory. The course of the degeneration process may vary among species, and may depend on the cause of deafness, but the present findings at least indicate that gradual retrograde degeneration of the auditory nerve is not an elemental process following severe damage to the organ of Corti.


Asunto(s)
Sordera/patología , Células Ciliadas Auditivas/ultraestructura , Degeneración Nerviosa , Ganglio Espiral de la Cóclea/ultraestructura , Animales , Umbral Auditivo , Sordera/inducido químicamente , Sordera/fisiopatología , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico , Furosemida , Cobayas , Kanamicina , Ototoxicidad , Ganglio Espiral de la Cóclea/fisiopatología
11.
J Int Adv Otol ; 16(1): 104-110, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32209520

RESUMEN

This study aims to obtain a better understanding of the number and distribution of spiral ganglion cell bodies (SGCBs) in the central modiolar trunk of the human cochlea with normal hearing as well as with hearing loss due to various pathological conditions. A detailed PubMed search was performed using the key words "human spiral ganglion cell population," "analysis of spiral ganglion cell population," "survival of human spiral ganglion cells," "human Rosenthal's canal," "human ganglion cell counts," and "distribution of human spiral ganglion cells" to identify articles published between 1931 and 2019. The articles were included if the number of SGCBs in the four segments of the human cochlea and angular depth distribution of the SGCBs were mentioned. Out of the 237 articles that were initially identified, 20 articles met the inclusion criteria. The presence of SGCBs inside the Rosenthal's canal (RC) in the modiolar trunk extended to an angular depth of 630°-680°, which is close to the end of the second turn of the cochlea. SGCBs in Segment-IV of the cochlea account for approximately 25-30% of the entire SGCB population, regardless of the cochlear condition (normal vs. pathologic). In normal-hearing subjects, the total number of SGCB cases ranged between 23,910 and 33,702; in patients with hearing loss, the same was between 5,733 and 28,220. This literature review elaborates on the current state of knowledge regarding the number and distribution of SGCBs in the human cochlea.


Asunto(s)
Cuerpo Celular/patología , Cóclea/patología , Pérdida Auditiva/patología , Ganglio Espiral de la Cóclea/patología , Adulto , Anciano , Anciano de 80 o más Años , Recuento de Células , Niño , Preescolar , Cóclea/anatomía & histología , Cóclea/fisiopatología , Pérdida Auditiva/fisiopatología , Pruebas Auditivas/métodos , Humanos , Lactante , Persona de Mediana Edad , Ganglio Espiral de la Cóclea/citología , Ganglio Espiral de la Cóclea/fisiopatología
12.
Int J Mol Med ; 45(6): 1851-1863, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32186779

RESUMEN

Sensorineural hearing loss (SNHL) is one of the major leading causes of hearing impairment, and is typically characterized by the degeneration of spiral ganglion neurons (SGNs). In previous studies by the authors, it was demonstrated that microRNA (miRNA or miR)­204­5p decreased the viability of SGNs by inhibiting the expression of transmembrane protease, serine 3 (TMPRSS3), which was closely associated with the development of SGNs. However, the upstream regulatory mechanism of miR­204­5p was not fully elucidated. The present study found that an important upstream regulatory factor of miR­204­5p, long non­coding RNA (lncRNA) EBLN3P, was expressed at low levels in impaired SGNs, whereas it was expressed at high levels in normal SGNs. Mechanistic analyses demonstrated that lncRNA EBLN3P functioned as a competing endogenous RNA (ceRNA) when regulating miR­204­5p in normal SGNs. In addition, lncRNA EBLN3P regulated TMPRSS3 expression via the regulation of miR­204­5p in normal SGNs. In vitro functional analysis revealed that lncRNA EBLN3P promoted the recovery of the viability of normal SGNs and inhibited the apoptosis of normal SGNs. Finally, the results revealed a recovery­promoting effect of lncRNA EBLN3P on the structure and function of impaired SGNs in models of deafness. On the whole, the findings of the present study demonstrate that lncRNA EBLN3P promotes the recovery of the function of impaired SGNs by competitively binding to miR­204­5p and regulating TMPRSS3 expression. This suggests that lncRNA EBLN3P may be a potential therapeutic target for diseases involving SNHL.


Asunto(s)
Proteínas de la Membrana/genética , MicroARNs/genética , Proteínas de Neoplasias/genética , Neuronas/fisiología , ARN Largo no Codificante/genética , Recuperación de la Función/genética , Serina Endopeptidasas/genética , Ganglio Espiral de la Cóclea/fisiopatología , Animales , Apoptosis/genética , Expresión Génica/genética , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/fisiopatología , Masculino , Ratones
13.
Hear Res ; 383: 107809, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31630082

RESUMEN

The electrically-evoked compound action potential (ECAP) is correlated with spiral ganglion neuron (SGN) density in cochlear implanted animals. In a previous study, we showed that ECAP amplitude growth function (AGF) linear slopes for stimuli with a constant interphase gap (IPG) changed significantly over time following implantation. Related studies have also shown that 1) IPG sensitivity for ECAP measures ("IPG Effect") is related to SGN density in animals and 2) the ECAP IPG Effect is related to speech recognition performance in humans with cochlear implants. The current study examined how the ECAP IPG Effect changed following cochlear implantation in four non-deafened guinea pigs with residual inner hair cells (IHCs) and 5 deafened, neurotrophin-treated guinea pigs. Simple impedances were measured on the same days as the ECAP measures. Generally, non-deafened implanted animals with higher SGN survival demonstrated higher ECAP AGF linear slope and peak amplitude values than the deafened, implanted guinea pigs. The ECAP IPG Effect for the AGF slopes and peak amplitudes was also larger in the hearing animals. The N1 latencies for a constant IPG were not different between groups, but the N1 latency IPG Effect was smaller in the non-deafened, implanted animals. Similar to previously reported results, ECAP measures using a fixed or changing IPG required as many as three months after implantation before a stable point could be calculated, but this was dependent on the animal and condition. For all ECAP measures most animals showed greater variance in the first 30 days post-implantation. Post-implantation changes in ECAPs and impedances were not correlated with one another. Results from this study are helpful for estimating the mechanisms underlying ECAP characteristics and have implications for clinical application of the ECAP measures in long-term human cochlear implant recipients. Specifically, these measures could help to monitor neural health over a period of time, or during a time of stability these measures could be used to help select electrode sites for activation in clinical programming.


Asunto(s)
Implantación Coclear/instrumentación , Implantes Cocleares , Sordera/rehabilitación , Potenciales Evocados , Ganglio Espiral de la Cóclea/fisiopatología , Estimulación Acústica , Animales , Muerte Celular , Sordera/patología , Sordera/fisiopatología , Sordera/psicología , Modelos Animales de Enfermedad , Estimulación Eléctrica , Cobayas , Tiempo de Reacción , Ganglio Espiral de la Cóclea/patología , Factores de Tiempo
14.
Audiol Neurootol ; 24(6): 271-278, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31665729

RESUMEN

BACKGROUND: Vestibular schwannoma (VS) is a benign intracranial neoplasm originating in the Schwann cells of the vestibular nerve. Despite its origin, the most common symptom is sensorineural hearing loss which is presented in more than 90% of patients. The underlying pathophysiology of this hearing loss has not been fully understood. OBJECTIVE: To assess the in vivo function of cochlear inner hair cells and spiral ganglion neurons in VS, cochlear dead regions (DRs) were evaluated via the threshold-equalizing noise (TEN) test in untreated VS patients. METHOD: Untreated patients diagnosed with sporadic unilateral VS and normal contralesional hearing were enrolled from July 2011 to June 2016. Audiometric evaluation including TEN tests were performed. Based on the magnetic resonance findings, characteristics of individual tumors were assessed. RESULTS: The average pure-tone threshold (word recognition score [WRS]) of 23 enrolled patients was 42.7 dB (76.1%). Nineteen DRs (11.8% of 161 tested frequencies) were found in 8 patients (34.8% of enrolled cases). Among the intracanalicular (IAC) tumors, 6 out of 10 ears (60%) carried DRs, while 2 of 13 (15.4%) showed DRs among the cerebellopontine angle (CPA) lesions (p = 0.039). Pure-tone thresholds and WRS were not different between the two groups. Logistic regression analysis showed that the tumor location, IAC versus CPA, was significantly associated with DRs (p = 0.041, Nagelkerke R2 = 0.471), whereas age, sex, tumor size, distance from the tumor to the cochlea, T2-weighted hypointensity on the MRI and pure-tone thresholds showed no significance. CONCLUSIONS: Cochlear DRs are detected in hearing losses associated with unilateral sporadic VS using the TEN test. Individual DRs were detected variously in high, mid, or low frequencies. In our preliminary data, IAC tumors showed a higher number of DRs than CPA tumors despite similar average hearing thresholds. Further studies including longitudinal follow-up of hearing as well as change in DRs may provide useful information about VS patients.


Asunto(s)
Cóclea/fisiopatología , Pérdida Auditiva Sensorineural/fisiopatología , Neuroma Acústico/fisiopatología , Ruido , Anciano , Audiometría de Tonos Puros , Umbral Auditivo , Femenino , Células Ciliadas Auditivas Internas/fisiología , Pérdida Auditiva/fisiopatología , Pérdida Auditiva Sensorineural/etiología , Pruebas Auditivas/métodos , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Neuroma Acústico/complicaciones , Neuroma Acústico/diagnóstico por imagen , Ganglio Espiral de la Cóclea/fisiopatología
15.
ORL J Otorhinolaryngol Relat Spec ; 81(2-3): 138-154, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31170714

RESUMEN

BACKGROUND/AIMS: Calcium-binding proteins in neurons buffer intracellular free Ca2+ ions, which interact with proteins controlling enzymatic and ion channel activity. The heterogeneous distribution of calretinin, calbindin, and parvalbumin influences calcium homeostasis, and calcium-related neuronal processes play an important role in neuronal aging and degeneration. This study evaluated age-related changes in calretinin, calbindin, and parvalbumin immune reactivity in spiral ganglion cells. METHODS: A total of 16 C57BL/6J and 16 129/SvJ mice at different ages (2, 4, 7, and 12 months) were included in the study. Hearing thresholds were assessed using auditory brainstem response before inner ears were excised for further evaluation. Semiquantitative immunohistochemistry for the aforementioned calcium-binding proteins was performed at the cellular level. RESULTS: The hearing thresholds of C57BL/6J and 129/SvJ mice increased significantly by 7 months of age. The average immune reactivity of calbin-din as well as the relative number of positive cells increased significantly with aging, but no significant alterations in calretinin or parvalbumin were observed. CONCLUSIONS: Upregulation of calbindin could serve as a protection to compensate for functional deficits that occur with aging. Expression of both calretinin and parvalbumin seem to be stabilizing factors in murine inner ears up to the age of 12 months in C57BL/6J and 129/SvJ mice.


Asunto(s)
Envejecimiento/genética , Calbindinas/genética , Regulación de la Expresión Génica , Pérdida Auditiva/genética , Ganglio Espiral de la Cóclea/metabolismo , Animales , Calbindinas/biosíntesis , Recuento de Células , Modelos Animales de Enfermedad , Femenino , Audición/fisiología , Pérdida Auditiva/metabolismo , Pérdida Auditiva/fisiopatología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ganglio Espiral de la Cóclea/patología , Ganglio Espiral de la Cóclea/fisiopatología
16.
Hear Res ; 376: 111-124, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30862414

RESUMEN

Strial dysfunction is commonly observed as a key consequence of aging in the cochlea. A large body of animal research, especially in the quiet-aged Mongolian gerbil, shows specific histopathological changes in the cochlear stria vascularis and the putatively corresponding effects on endocochlear potential and auditory nerve responses. However, recent work suggests that synaptopathy, or the loss of inner hair cell-auditory nerve fiber synapses, also presents as a consequence of aging. It is now believed that the loss of synapses is the earliest age-related degenerative event. The present review aims to integrate classic and novel research on age-related pathologies of the inner ear. First, we summarize current knowledge on age-related strial dysfunction and synaptopathy. We describe how these cochlear pathologies fit into the categories for presbyacusis, as first defined by Schuknecht in the '70s. Further, we discuss how strial dysfunction and synaptopathy affect sound coding by the auditory nerve and how they can be experimentally induced to study their specific contributions to age-related hearing deficits. As such, we aim to give an overview of the current literature on age-related cochlear pathologies and hope to inspire further research on the role of cochlear aging in age-related hearing deficits.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Cóclea/patología , Cóclea/fisiopatología , Animales , Cóclea/inervación , Nervio Coclear/fisiopatología , Modelos Animales de Enfermedad , Endolinfa/metabolismo , Gerbillinae , Humanos , Modelos Biológicos , Potasio/metabolismo , Presbiacusia/etiología , Presbiacusia/patología , Presbiacusia/fisiopatología , Ganglio Espiral de la Cóclea/patología , Ganglio Espiral de la Cóclea/fisiopatología , Estría Vascular/patología , Estría Vascular/fisiología , Sinapsis/patología
17.
Adv Exp Med Biol ; 1130: 93-107, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30915703

RESUMEN

In the auditory system, the primary sensory neurons, spiral ganglion neurons (SGNs), transmit complex acoustic information from hair cells to the second-order sensory neurons in the cochlear nucleus for sound processing, thus building the initial bridge between the physical world of sound and the perception of that sound. Cochlear SGN loss causes irreversible hearing impairment because this type of neural cell cannot regenerate. A better understanding of the molecular mechanisms of formation, structure, degeneration, and protection of SGNs will help to design potential therapeutic strategies for preservation and replacement of them in the cochlear implant recipient. In this review, we described and summarized the following about SGNs: (1) their cell biology and their peripheral and central connections, (2) mechanisms of their neuronal damage and their protection, and (3) the neural and synaptic mechanism of auditory neuropathy and current options for hearing rehabilitation from auditory neuropathy. The updates of the research progress and the significant issues on these topics were discussed.


Asunto(s)
Células Ciliadas Auditivas/citología , Pérdida Auditiva Central/prevención & control , Neuronas/citología , Ganglio Espiral de la Cóclea/fisiopatología , Humanos
18.
Biomed Res Int ; 2019: 3626032, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30766882

RESUMEN

Lead exposure causes or aggravates hearing damage to human or animal, but the detailed effects of lead exposure on auditory system including injury sites of the cochlea in mammal remain controversy. To investigate the effect of chronic lead exposure on auditory system, 40 adult guinea pigs with normal hearing were randomly divided into five groups. They were fed 2 mmol/L lead acetate in drinking water for 0, 15, 30, 60, and 90 days (n = 8), respectively. Lead concentrations in blood, cochlea, and brainstem were measured. Auditory function was measured by auditory brainstem response (ABR) and distortion product otoacoustic emission (DPOAE). The morphology of cochlea and brainstem was observed, and expression of autophagy-related protein in brainstem was also assessed. The blood lead concentration reached a high level at the 15th day and kept stable, but the lead level in brainstem and cochlear tissue increased obviously at the 60th day and 90th day of lead exposure, respectively. There was no significant difference in the morphology of hair cells and stria vascularis (SV) among these five groups, but the number of spiral ganglion neuron (SGN) gradually decreased after 60 days. The differences of ABR thresholds and DPOAE amplitudes were not statistically significant among each group, but I wave latency, III latency, and I-III wave interval of ABR were delayed with the prolonging of time of lead exposure. The expressions of autophagy-related protein ATG5, ATG6, and LC3B in brainstem were increased after 30 days. These results suggest that the key target of lead toxicity was the auditory nerve conduction pathway including SGNs and brainstem, rather than cochlear hair cells and SV. Autophagy may play a very important role in lead toxicity to auditory nervous system.


Asunto(s)
Cóclea/efectos de los fármacos , Audición/efectos de los fármacos , Plomo/efectos adversos , Síndromes de Neurotoxicidad/fisiopatología , Animales , Umbral Auditivo/efectos de los fármacos , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/fisiopatología , Cóclea/fisiopatología , Nervio Coclear/efectos de los fármacos , Nervio Coclear/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Femenino , Cobayas , Células Ciliadas Auditivas/efectos de los fármacos , Plomo/sangre , Masculino , Neuronas/efectos de los fármacos , Emisiones Otoacústicas Espontáneas/efectos de los fármacos , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/fisiopatología , Estría Vascular/efectos de los fármacos
19.
Neuroscience ; 406: 345-355, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30742960

RESUMEN

Conductive hearing loss is a prevalent condition globally. It remains unclear whether conductive hearing loss that occurs during early development disrupts auditory peripheral systems. In this study, a mouse model of conductive auditory deprivation (CAD) was achieved using external auditory canal closure on postnatal day 12, which marks the onset of external ear canal opening. Short-term (2 weeks) and long-term (6 weeks) deprivations involving external ear canal closure were conducted. Mice were examined immediately, 4 weeks, and 8 weeks after deprivation. Short-term deprivation induced reversible auditory brainstem response (ABR) threshold and latencies of ABR wave I, whereas long-term deprivation caused irreversible ABR thresholds and latencies of ABR wave I. Complete recovery of ribbon synapses and latencies of ABR wave I was observed in the short-term group. In contrast, we observed irreversible ABR thresholds, latencies of ABR wave I, and quantity of ribbon synapses in the long-term deprivation group. Positive 8-hydroxy-2'-deoxyguanosine signals were noted in cochlear hair cells in the long-term group, suggesting that long-term auditory deprivation could disrupt auditory maturation via mitochondrial damage in cochlear hair cells. Conversely, no significant changes in cellular morphology were observed in cochlear hair cells and spiral ganglion cells in either short- or long-term groups. Collectively, our findings suggest that long-term conductive hearing deprivation during early stages of auditory development can cause significant and irreversible disruption that persists into adulthood.


Asunto(s)
Umbral Auditivo/fisiología , Cóclea/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Animales , Audición/fisiología , Ratones Endogámicos C57BL , Ganglio Espiral de la Cóclea/fisiopatología , Sinapsis/fisiología , Tiempo
20.
J Acoust Soc Am ; 144(5): 2751, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30522299

RESUMEN

The symmetric biphasic pulses used in contemporary cochlear implants (CIs) consist of both cathodic and anodic currents, which may stimulate different sites on spiral ganglion neurons and, potentially, interact with each other. The effect on the order of anodic and cathodic stimulation on loudness at short inter-pulse intervals (IPIs; 0-800 µs) is investigated. Pairs of opposite-polarity pseudomonophasic (PS) pulses were used and the amplitude of each pulse was manipulated independently. In experiment 1 the two PS pulses differed in their current level in order to elicit the same loudness when presented separately. Six users of the Advanced Bionics CI (Valencia, CA) loudness-ranked trains of the pulse pairs using a midpoint-comparison procedure. Stimuli with anodic-leading polarity were louder than those with cathodic-leading polarity for IPIs shorter than 400 µs. This effect was small-about 0.3 dB-but consistent across listeners. When the same procedure was repeated with both PS pulses having the same current level (experiment 2), anodic-leading stimuli were still louder than cathodic-leading stimuli at very short intervals. However, when using symmetric biphasic pulses (experiment 3) the effect disappeared at short intervals and reversed at long intervals. Possible peripheral sources of such polarity interactions are discussed.


Asunto(s)
Percepción Auditiva/fisiología , Implantes Cocleares/efectos adversos , Percepción Sonora/fisiología , Ganglio Espiral de la Cóclea/fisiopatología , Estimulación Acústica , Anciano , Implantación Coclear/métodos , Implantes Cocleares/estadística & datos numéricos , Estimulación Eléctrica/efectos adversos , Electrodos Implantados/normas , Humanos , Persona de Mediana Edad , Discriminación de la Altura Tonal/fisiología , Diseño de Prótesis , Ganglio Espiral de la Cóclea/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA